1
|
Oh N, Hwang J, Kang MS, Yoo CY, Kwak M, Han DW. Versatile and Marvelous Potentials of Polydeoxyribonucleotide for Tissue Engineering and Regeneration. Biomater Res 2025; 29:0183. [PMID: 40231205 PMCID: PMC11994882 DOI: 10.34133/bmr.0183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/10/2025] [Accepted: 03/15/2025] [Indexed: 04/16/2025] Open
Abstract
Over the past decade, substantial focus has been placed on polydeoxyribonucleotide (PDRN) due to its promising pharmacological properties, making it a valuable candidate for tissue engineering applications. Accordingly, this paper aims to review and summarize the latest experimental research on PDRN in the context of tissue engineering and regeneration. The unique biochemical mechanisms of PDRN to promote cellular behavior and regeneration are summarized. We categorize commonly utilized PDRN-based tissue engineering fields as neuromuscular tissues, diabetic wound or skin, and bone regeneration. At the same time, we explore scaffold strategies for integrating PDRN into bioceramics, polymers, and cell/tissue-derived materials, along with its combination with photo/electromodulation techniques. Furthermore, we discuss potential opportunities and challenges in translating PDRN-based approaches into clinical practice. We expect future interdisciplinary research and clinical trials to evaluate the long-term efficacy and safety of PDRN while emphasizing standardization and quality control to ensure its consistency and effectiveness in regenerative applications.
Collapse
Affiliation(s)
- Nuri Oh
- Department of Chemistry and Biology,
Korea Science Academy of Korea Advanced Institute of Science and Technology, Busan 47162, Republic of Korea
| | - Juyoung Hwang
- Department of Chemistry,
Pukyong National University, Busan 48513, Republic of Korea
- Smart Gym-Based Translational Research Center for Active Senior’s Healthcare,
Pukyong National University, Busan 48513, Republic of Korea
- Ajou Energy Science Research Center,
Ajou University, Suwon 16499, Republic of Korea
| | - Moon Sung Kang
- Research Institute of Mechanical Technology,
Pusan National University, Busan 46241, Republic of Korea
| | - Chung-Yul Yoo
- Department of Energy Systems Research and Chemistry,
Ajou University, Suwon 16499, Republic of Korea
| | - Minseok Kwak
- Department of Chemistry,
Pukyong National University, Busan 48513, Republic of Korea
- Smart Gym-Based Translational Research Center for Active Senior’s Healthcare,
Pukyong National University, Busan 48513, Republic of Korea
- Industry 4.0 Convergence Bionics Engineering,
Pukyong National University, Busan 48513, Republic of Korea
| | - Dong-Wook Han
- Department of Cogno-Mechatronics Engineering,
Pusan National University, Busan 46241, Republic of Korea
- Institute of Nano-Bio Convergence,
Pusan National University, Busan 46241, Republic of Korea
| |
Collapse
|
2
|
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
|
3
|
Zhang M, Mi M, Hu Z, Li L, Chen Z, Gao X, Liu D, Xu B, Liu Y. Polydopamine-Based Biomaterials in Orthopedic Therapeutics: Properties, Applications, and Future Perspectives. Drug Des Devel Ther 2024; 18:3765-3790. [PMID: 39219693 PMCID: PMC11363944 DOI: 10.2147/dddt.s473007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 08/10/2024] [Indexed: 09/04/2024] Open
Abstract
Polydopamine is a versatile and modifiable polymer, known for its excellent biocompatibility and adhesiveness. It can also be engineered into a variety of nanoparticles and biomaterials for drug delivery, functional modification, making it an excellent choice to enhance the prevention and treatment of orthopedic diseases. Currently, the application of polydopamine biomaterials in orthopedic disease prevention and treatment is in its early stages, despite some initial achievements. This article aims to review these applications to encourage further development of polydopamine for orthopedic therapeutic needs. We detail the properties of polydopamine and its biomaterial types, highlighting its superior performance in functional modification on nanoparticles and materials. Additionally, we also explore the challenges and future prospects in developing optimal polydopamine biomaterials for clinical use in orthopedic disease prevention and treatment.
Collapse
Affiliation(s)
- Min Zhang
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, 524037, People’s Republic of China
- Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Provincial Administration of Traditional Chinese Medicine (Central People’s Hospital of Zhanjiang), Zhanjiang, 524037, People’s Republic of China
- Marine Medical Research Institute of Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, 524023, People’s Republic of China
| | - Man Mi
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, 524037, People’s Republic of China
- Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Provincial Administration of Traditional Chinese Medicine (Central People’s Hospital of Zhanjiang), Zhanjiang, 524037, People’s Republic of China
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang, 524023, People’s Republic of China
| | - Zilong Hu
- Marine Medical Research Institute of Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, 524023, People’s Republic of China
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang, 524023, People’s Republic of China
| | - Lixian Li
- Marine Medical Research Institute of Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, 524023, People’s Republic of China
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang, 524023, People’s Republic of China
| | - Zhiping Chen
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, 524037, People’s Republic of China
- Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Provincial Administration of Traditional Chinese Medicine (Central People’s Hospital of Zhanjiang), Zhanjiang, 524037, People’s Republic of China
- Marine Medical Research Institute of Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, 524023, People’s Republic of China
| | - Xiang Gao
- Stem Cell Research and Cellular Therapy Center, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, People’s Republic of China
| | - Di Liu
- Marine Medical Research Institute of Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, 524023, People’s Republic of China
- Guangdong Provincial Key Laboratory for Research and Development of Natural Drug, School of Pharmacy, Guangdong Medical University, Zhanjiang, 524023, People’s Republic of China
| | - Bilian Xu
- Marine Medical Research Institute of Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, 524023, People’s Republic of China
| | - Yanzhi Liu
- Zhanjiang Key Laboratory of Orthopaedic Technology and Trauma Treatment, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, 524037, People’s Republic of China
- Key Laboratory of Traditional Chinese Medicine for the Prevention and Treatment of Infectious Diseases, Guangdong Provincial Administration of Traditional Chinese Medicine (Central People’s Hospital of Zhanjiang), Zhanjiang, 524037, People’s Republic of China
- Marine Medical Research Institute of Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, 524023, People’s Republic of China
| |
Collapse
|
4
|
Lee JK, Kim DS, Park SY, Jung JW, Baek SW, Lee S, Kim JH, Ahn TK, Han DK. Osteoporotic Bone Regeneration via Plenished Biomimetic PLGA Scaffold with Sequential Release System. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310734. [PMID: 38143290 DOI: 10.1002/smll.202310734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 12/21/2023] [Indexed: 12/26/2023]
Abstract
Achieving satisfactory bone tissue regeneration in osteoporotic patients with ordinary biomaterials is challenging because of the decreased bone mineral density and aberrant bone microenvironment. In addressing this issue, a biomimetic scaffold (PMEH/SP), incorporating 4-hexylresorcinol (4HR), and substance P (SP) into the poly(lactic-go-glycolic acid) (PLGA) scaffold with magnesium hydroxide (M) and extracellular matrix (E) is introduced, enabling the consecutive release of bioactive agents. 4HR and SP induced the phosphorylation of p38 MAPK and ERK in human umbilical vein endothelial cells (HUVECs), thereby upregulating VEGF expression level. The migration and tube-forming ability of endothelial cells can be promoted by the scaffold, which accelerates the formation and maturation of the bone. Moreover, 4HR played a crucial role in the inhibition of osteoclastogenesis by interrupting the IκB/NF-κB signaling pathway and exhibiting SP, thereby enhancing the migration and angiogenesis of HUVECs. Based on such a synergistic effect, osteoporosis can be suppressed, and bone regeneration can be achieved by inhibiting the RANKL pathway in vitro and in vivo, which is a commonly known mechanism of bone physiology. Therefore, the study presents a promising approach for developing a multifunctional regenerative material for sophisticated osteoporotic bone regeneration.
Collapse
Affiliation(s)
- Jun-Kyu Lee
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Da-Seul Kim
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women's Hospital, Cambridge, MA, 02139, USA
| | - So-Yeon Park
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Ji-Won Jung
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Seung-Woon Baek
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Semi Lee
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Jun Hyuk Kim
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Tae-Keun Ahn
- Department of Orthopedic Surgery, CHA Bundang Medical Center, CHA University, Seongnam-si, Gyeonggi-do, 13496, Republic of Korea
| | - Dong Keun Han
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| |
Collapse
|
5
|
Morse KW, Sun J, Hu L, Bok S, Debnath S, Cung M, Yallowitz AR, Meyers KN, Iyer S, Greenblatt MB. Development of Murine Anterior Interbody and Posterolateral Spinal Fusion Techniques. J Bone Joint Surg Am 2024; 106:735-745. [PMID: 38194481 DOI: 10.2106/jbjs.23.00690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2024]
Abstract
BACKGROUND Multiple animal models have previously been utilized to investigate anterior fusion techniques, but a mouse model has yet to be developed. The purpose of this study was to develop murine anterior interbody and posterolateral fusion techniques. METHODS Mice underwent either anterior interbody or posterolateral spinal fusion. A protocol was developed for both procedures, including a description of the relevant anatomy. Samples were subjected to micro-computed tomography to assess fusion success and underwent biomechanical testing with use of 4-point bending. Lastly, samples were fixed and embedded for histologic evaluation. RESULTS Surgical techniques for anterior interbody and posterolateral fusion were developed. The fusion rate was 83.3% in the anterior interbody model and 100% in the posterolateral model. Compared with a control, the posterolateral model exhibited a greater elastic modulus. Histologic analysis demonstrated endochondral ossification between bridging segments, further confirming the fusion efficacy in both models. CONCLUSIONS The murine anterior interbody and posterolateral fusion models are efficacious and provide an ideal platform for studying the molecular and cellular mechanisms mediating spinal fusion. CLINICAL RELEVANCE Given the extensive genetic tools available in murine disease models, use of fusion models such as ours can enable determination of the underlying genetic pathways involved in spinal fusion.
Collapse
Affiliation(s)
- Kyle W Morse
- Department of Spine Surgery, Hospital for Special Surgery, New York, NY
| | - Jun Sun
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY
| | - Lingling Hu
- Department of Spine Surgery, Hospital for Special Surgery, New York, NY
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY
- Research Division, Hospital for Special Surgery, New York, NY
| | - Seoyeon Bok
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY
| | - Shawon Debnath
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY
| | - Michelle Cung
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY
| | - Alisha R Yallowitz
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY
| | - Kathleen N Meyers
- Department of Biomechanics, Hospital for Special Surgery, New York, NY
| | - Sravisht Iyer
- Department of Spine Surgery, Hospital for Special Surgery, New York, NY
| | - Matthew B Greenblatt
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY
- Research Division, Hospital for Special Surgery, New York, NY
| |
Collapse
|
6
|
Xiang S, Zhang C, Guan Z, Li X, Liu Y, Feng G, Luo X, Zhang B, Weng J, Xiao D. Preparation of a novel antibacterial magnesium carbonate coating on a titanium surface and its in vitro biocompatibility. RSC Adv 2024; 14:10516-10525. [PMID: 38567331 PMCID: PMC10985587 DOI: 10.1039/d4ra00399c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 03/25/2024] [Indexed: 04/04/2024] Open
Abstract
Magnesium-based coatings have attracted great attention in surface modification of titanium implants due to their superior angiogenic and osteogenic properties. However, their biological effects as a carbonate-based constituent remain unrevealed. In this study, magnesium carbonate coatings were prepared on titanium surfaces under hydrothermal conditions and subsequently treated with hydrogen peroxide. Also, their antibacterial activity and in vitro cell biocompatibility were evaluated. The obtained coatings consisted of nanoparticles without cracks and exhibited excellent adhesion to the substrate. X-ray diffraction (XRD) results indicated pure magnesium carbonate coatings formed on the Ti surface after hydrothermal treatment. After hydrogen peroxide treatment, the phase composition of the coatings had no obvious change. Compared to the untreated coatings, the hydrogen peroxide-treated coatings showed increased surface roughness and hydrophilicity. Co-culture with Staphylococcus aureus (S. aureus) demonstrated that the obtained coatings had good antibacterial activity. In vitro cell culture results showed that the hydrogen peroxide-treated coatings enhanced the viability, proliferation, and osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs). These findings suggest that this MgCO3-based coating exhibits excellent antibacterial performance and osteogenic potential. Based on the above, this study provides a simple method for preparing titanium implants with dual antibacterial and osteogenic capabilities, holding great promise in clinical applications.
Collapse
Affiliation(s)
- Shougang Xiang
- Department of Orthopaedics, Research Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital (Beijing Anzhen Hospital Nanchong Hospital), The Second Clinical College of North Sichuan Medical College Nanchong Sichuan 637000 China
| | - Chengdong Zhang
- Key Laboratory of Advanced Technologies of Materials (MOE), School of Materials Science and Engineering, Southwest Jiaotong University Chengdu Sichuan 610031 China
| | - Zhenju Guan
- Department of Orthopaedics, Research Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital (Beijing Anzhen Hospital Nanchong Hospital), The Second Clinical College of North Sichuan Medical College Nanchong Sichuan 637000 China
| | - Xingping Li
- Department of Orthopaedics, Chengfei Hospital Chengdu Sichuan 610091 China
| | - Yumei Liu
- Collaboration Innovation Center for Tissue Repair Material Engineering Technology, China West Normal University Nanchong Sichuan 637002 China
| | - Gang Feng
- Department of Orthopaedics, Research Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital (Beijing Anzhen Hospital Nanchong Hospital), The Second Clinical College of North Sichuan Medical College Nanchong Sichuan 637000 China
| | - Xuwei Luo
- Department of Orthopaedics, Research Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital (Beijing Anzhen Hospital Nanchong Hospital), The Second Clinical College of North Sichuan Medical College Nanchong Sichuan 637000 China
| | - Bo Zhang
- Department of Orthopaedics, Research Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital (Beijing Anzhen Hospital Nanchong Hospital), The Second Clinical College of North Sichuan Medical College Nanchong Sichuan 637000 China
| | - Jie Weng
- Key Laboratory of Advanced Technologies of Materials (MOE), School of Materials Science and Engineering, Southwest Jiaotong University Chengdu Sichuan 610031 China
| | - Dongqin Xiao
- Department of Orthopaedics, Research Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital (Beijing Anzhen Hospital Nanchong Hospital), The Second Clinical College of North Sichuan Medical College Nanchong Sichuan 637000 China
| |
Collapse
|
7
|
Lee S, Baek SW, Kim DS, Park SY, Kim JH, Jung JW, Lee JK, Park GM, Park CG, Han DK. Injectable Microparticle-containing hydrogel with controlled release of bioactive molecules for facial rejuvenation. Mater Today Bio 2024; 24:100890. [PMID: 38162281 PMCID: PMC10755792 DOI: 10.1016/j.mtbio.2023.100890] [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: 10/18/2023] [Revised: 11/20/2023] [Accepted: 11/28/2023] [Indexed: 01/03/2024] Open
Abstract
The skin is the largest organ and a crucial barrier for protection against various intrinsic and extrinsic factors. As we age, the skin's components become more vulnerable to damage, forming wrinkles. Among different procedures, hyaluronic acid-based hydrogel has been extensively utilized for skin regeneration and reducing wrinkles. However, it has limitations like low retention and weak mechanical properties. In this study, we suggested the poly(l-lactic acid) (PLLA) microparticles containing alkaline magnesium hydroxide and nitric oxide-generating zinc oxide and rejuvenative hyaluronic acid (HA) hydrogels including these functional microparticles and asiaticoside, creating a novel delivery system for skin rejuvenation and regeneration. The fabricated rejuvenative hydrogels have exhibited enhanced biocompatibility, pH neutralization, reactive oxygen species scavenging, collagen biosynthesis, and angiogenesis capabilities in vitro and in vivo. Additionally, an excellent volume retention ability was demonstrated due to the numerous hydrogen bonds that formed between hyaluronic acid and asiaticoside. Overall, our advanced injectable hydrogel containing functional microparticles, with controlled release of bioactive molecules, has a significant potential for enhancing the regeneration and rejuvenation of the skin.
Collapse
Affiliation(s)
- Semi Lee
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyenggi 13488, South Korea
| | - Seung-Woon Baek
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyenggi 13488, South Korea
- Department of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyenggi 16419, South Korea
- Department of Intelligent Precision Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyenggi 16419, South Korea
| | - Da-Seul Kim
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyenggi 13488, South Korea
| | - So-Yeon Park
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyenggi 13488, South Korea
- Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, South Korea
| | - Jun Hyuk Kim
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyenggi 13488, South Korea
| | - Ji-Won Jung
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyenggi 13488, South Korea
| | - Jun-Kyu Lee
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyenggi 13488, South Korea
| | - Gi-Min Park
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyenggi 13488, South Korea
| | - Chun Gwon Park
- Department of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyenggi 16419, South Korea
- Department of Intelligent Precision Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyenggi 16419, South Korea
| | - Dong Keun Han
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyenggi 13488, South Korea
| |
Collapse
|
8
|
Sharma A, Krishnan M, Ganganahalli G, Saraswathy S, Johnson R, Iyer SR. Microarray illustrates enhanced mechanistic action towards osteogenesis for magnesium aluminate spinel ceramic-based polyphasic composite scaffold with mesenchymal stem cells and bone morphogenetic protein 2. J Biomed Mater Res B Appl Biomater 2023; 111:1858-1868. [PMID: 35289496 DOI: 10.1002/jbm.b.35051] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 02/24/2022] [Accepted: 03/03/2022] [Indexed: 11/10/2022]
Abstract
Spinel (magnesium aluminate MgAl2 O4 ) ceramic-based polyphasic composite scaffold has been recently reported for craniofacial bone tissue engineering. Improving the osteogenic effects of such composite scaffolds with bone morphogenetic proteins (BMP2) is an intensely researched area. This study investigated the gene interactions of this scaffold with BMP2 and mesenchymal stem cells (MSCs). Human bone marrow MSCs were cultured in 3 groups: Group 1-Control (BMSCs), Group 2-BMSC with BMP2, and Group 3-BMSC with scaffold and BMP2. After RNA isolation, gene expression analysis was done by microarray. Differentially expressed genes (DEGs) (-1.0 > fold changes>1 and p value <.05) were studied for their function and gene ontologies using Database for Annotation, Visualization and Integrated Discovery (DAVID). They were further studied by protein-protein interaction network analysis using STRING and MCODE Cytoscape plugin database. Group 3 showed up regulation of 3222 genes against 2158 of Group 2. Group 3 had five annotation clusters with enrichment scores from 2.08 to 3.93. Group 2 had only one cluster. Group 3 showed activation of all major osteogenic pathways: TGF, BMP2, WNT, SMAD, and Notch gene signaling with effects of calcium and magnesium released from the scaffold. Downstream effect of all these caused significant activation of RUNX2, the key transcriptional regulator of osteogenesis in Group 3. STRING and MCODE Cytoscape plugin demonstrated the interactions. The enhanced MSC differentiation for osteogenesis with the addition of BMP2 to the polyphasic composite scaffold proposed promising clinical applications for bone tissue engineering.
Collapse
Affiliation(s)
- Anu Sharma
- Department of Dental Research and Implantology, Institute of Nuclear Medicine and Allied Sciences (INMAS), Defence Research and Development Organization (DRDO), Delhi, India
| | - Manu Krishnan
- Department of Dental Research and Implantology, Institute of Nuclear Medicine and Allied Sciences (INMAS), Defence Research and Development Organization (DRDO), Delhi, India
| | - Gurudatta Ganganahalli
- Division of Stem Cell and Gene Therapy Research, Institute of Nuclear Medicine and Allied Sciences (INMAS), Defence Research and Development Organization (DRDO), Delhi, India
| | - Seema Saraswathy
- Department of Biochemistry, Army College of Medical Sciences (ACMS), Delhi, India
| | - Roy Johnson
- Centre for Ceramic Processing, International Advanced Research Centre for Powder Metallurgy and New Materials (ARCI), Hyderabad, India
| | - Satish R Iyer
- Directorate General of Dental Services (DGDS), Delhi, India
| |
Collapse
|
9
|
Emadi H, Karevan M, Masoudi Rad M, Sadeghzade S, Pahlevanzadeh F, Khodaei M, Khayatzadeh S, Lotfian S. Bioactive and Biodegradable Polycaprolactone-Based Nanocomposite for Bone Repair Applications. Polymers (Basel) 2023; 15:3617. [PMID: 37688243 PMCID: PMC10490551 DOI: 10.3390/polym15173617] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/07/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
This study investigated the relationship between the structure and mechanical properties of polycaprolactone (PCL) nanocomposites reinforced with baghdadite, a newly introduced bioactive agent. The baghdadite nanoparticles were synthesised using the sol-gel method and incorporated into PCL films using the solvent casting technique. The results showed that adding baghdadite to PCL improved the nanocomposites' tensile strength and elastic modulus, consistent with the results obtained from the prediction models of mechanical properties. The tensile strength increased from 16 to 21 MPa, and the elastic modulus enhanced from 149 to 194 MPa with fillers compared to test specimens without fillers. The thermal properties of the nanocomposites were also improved, with the degradation temperature increasing from 388 °C to 402 °C when 10% baghdadite was added to PCL. Furthermore, it was found that the nanocomposites containing baghdadite showed an apatite-like layer on their surfaces when exposed to simulated body solution (SBF) for 28 days, especially in the film containing 20% nanoparticles (PB20), which exhibited higher apatite density. The addition of baghdadite nanoparticles into pure PCL also improved the viability of MG63 cells, increasing the viability percentage on day five from 103 in PCL to 136 in PB20. Additionally, PB20 showed a favourable degradation rate in PBS solution, increasing mass loss from 2.63 to 4.08 per cent over four weeks. Overall, this study provides valuable insights into the structure-property relationships of biodegradable-bioactive nanocomposites, particularly those reinforced with new bioactive agents.
Collapse
Affiliation(s)
- Hosein Emadi
- School of Mechanical Engineering, College of Engineering, University of Tehran, Tehran 14176-14411, Iran
- Department of Mechanical Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran;
| | - Mehdi Karevan
- Department of Mechanical Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran;
| | - Maryam Masoudi Rad
- Department of Chemical Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran;
| | - Sorour Sadeghzade
- Shenzhen Key Laboratory of Soft Mechanics & Smart Manufacturing, Department of Mechanics and Aerospace Engineering, Southern University of Science and Technology, Shenzhen 518055, China;
| | - Farnoosh Pahlevanzadeh
- Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran;
| | - Mohammad Khodaei
- Materials Engineering Group, Golpayegan College of Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran;
| | - Saber Khayatzadeh
- Department of Design and Mathematics, University of the West of England, Bristol BS16 1QY, UK
| | - Saeid Lotfian
- Faculty of Engineering, University of Strathclyde, Glasgow G4 0LZ, UK
| |
Collapse
|
10
|
de Jesús Martín-Camacho U, Rodríguez-Barajas N, Alberto Sánchez-Burgos J, Pérez-Larios A. Weibull β value for the discernment of drug release mechanism of PLGA particles. Int J Pharm 2023; 640:123017. [PMID: 37149112 DOI: 10.1016/j.ijpharm.2023.123017] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 04/27/2023] [Accepted: 04/30/2023] [Indexed: 05/08/2023]
Abstract
Mathematical models are used to characterize and optimize drug release in drug delivery systems (DDS). One of the most widely used DDS is the poly(lactic-co-glycolic acid) (PLGA)-based polymeric matrix owing to its biodegradability, biocompatibility, and easy manipulation of its properties through the manipulation of synthesis processes. Over the years, the Korsmeyer-Peppas model has been the most widely used model for characterizing the release profiles of PLGA DDS. However, owing to the limitations of the Korsmeyer-Peppas model, the Weibull model has emerged as an alternative for the characterization of the release profiles of PLGA polymeric matrices. The purpose of this study was to establish a correlation between the n and β parameters of the Korsmeyer-Peppas and Weibull models and to use the Weibull model to discern the drug release mechanism. A total of 451 datasets describing the overtime drug release of PLGA-based formulations from 173 scientific articles were fitted to both models. The Korsmeyer-Peppas model had a mean Akaike Information Criteria (AIC) value of 54.52 and an n value of 0.42, while the Weibull model had a mean AIC of 51.99 and a β value of 0.55, and by using reduced major axis regression values, a high correlation was found between the n and β values. These results demonstrate the ability of the Weibull model to characterize the release profiles of PLGA-based matrices and the usefulness of the β parameter for determining the drug release mechanism.
Collapse
Affiliation(s)
- Ubaldo de Jesús Martín-Camacho
- Laboratorio de Investigación en Materiales, Agua y Energía, Departamento de Ingeniería, Centro Universitario de los Altos, Universidad de Guadalajara, Tepatitlán de Morelos, Jal., México, 47600
| | - Noé Rodríguez-Barajas
- Laboratorio de Investigación en Materiales, Agua y Energía, Departamento de Ingeniería, Centro Universitario de los Altos, Universidad de Guadalajara, Tepatitlán de Morelos, Jal., México, 47600
| | | | - Alejandro Pérez-Larios
- Laboratorio de Investigación en Materiales, Agua y Energía, Departamento de Ingeniería, Centro Universitario de los Altos, Universidad de Guadalajara, Tepatitlán de Morelos, Jal., México, 47600.
| |
Collapse
|
11
|
Lee HY, Kim DS, Hwang GY, Lee JK, Lee HL, Jung JW, Hwang SY, Baek SW, Yoon SL, Ha Y, Kim KN, Han I, Han DK, Lee CK. Multi-modulation of immune-inflammatory response using bioactive molecule-integrated PLGA composite for spinal fusion. Mater Today Bio 2023; 19:100611. [PMID: 36969699 PMCID: PMC10034518 DOI: 10.1016/j.mtbio.2023.100611] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/03/2023] [Accepted: 03/11/2023] [Indexed: 03/15/2023] Open
Abstract
Despite current developments in bone substitute technology for spinal fusion, there is a lack of adequate materials for bone regeneration in clinical applications. Recombinant human bone morphogenetic protein-2 (rhBMP-2) is commercially available, but a severe inflammatory response is a known side effect. Bone graft substitutes that enhance osteogenesis without adverse effects are needed. We developed a bioactive molecule-laden PLGA composite with multi-modulation for bone fusion. This bioresorbable composite scaffold was considered for bone tissue engineering. Among the main components, magnesium hydroxide (MH) aids in reduction of acute inflammation affecting disruption of new bone formation. Decellularized bone extracellular matrix (bECM) and demineralized bone matrix (DBM) composites were used for osteoconductive and osteoinductive activities. A bioactive molecule, polydeoxyribonucleotide (PDRN, PN), derived from trout was used for angiogenesis during bone regeneration. A nano-emulsion method that included Span 80 was used to fabricate bioactive PLGA-MH-bECM/DBM-PDRN (PME2/PN) composite to obtain a highly effective and safe scaffold. The synergistic effect provided by PME2/PN improved not only osteogenic and angiogenic gene expression for bone fusion but also improved immunosuppression and polarization of macrophages that were important for bone tissue repair, using a rat model of posterolateral spinal fusion (PLF). It thus had sufficient biocompatibility and bioactivity for spinal fusion.
Collapse
Affiliation(s)
- Hye Yeong Lee
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
| | - Da-Seul Kim
- Department of Biomedical Science, CHA University, Gyeonggi-do, 13488, Republic of Korea
- School of Integrative Engineering, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Gwang Yong Hwang
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
| | - Jun-Kyu Lee
- Department of Biomedical Science, CHA University, Gyeonggi-do, 13488, Republic of Korea
| | - Hye-Lan Lee
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
| | - Ji-Won Jung
- Department of Biomedical Science, CHA University, Gyeonggi-do, 13488, Republic of Korea
| | - Sae Yeon Hwang
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
- Graduate Program in Bioindustrial Engineering, Yonsei University, Seoul, 03722, Republic of Korea
| | - Seung-Woon Baek
- Department of Biomedical Science, CHA University, Gyeonggi-do, 13488, 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
| | - Keung Nyun Kim
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
| | - Inbo Han
- Department of Neurosurgery, CHA University School of Medicine, CHA Bungdang Medical Center, Gyeonggi-do, 13496, Republic of Korea
| | - Dong Keun Han
- Department of Biomedical Science, CHA University, Gyeonggi-do, 13488, Republic of Korea
- Corresponding author.
| | - Chang Kyu Lee
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, Seoul, 03722, Republic of Korea
- Corresponding author.
| |
Collapse
|
12
|
Lee J, Kim D, Park S, Baek S, Jung J, Kim T, Han DK. Nitric Oxide-Releasing Bioinspired Scaffold for Exquisite Regeneration of Osteoporotic Bone via Regulation of Homeostasis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205336. [PMID: 36581472 PMCID: PMC9951336 DOI: 10.1002/advs.202205336] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/14/2022] [Indexed: 06/17/2023]
Abstract
Osteoporotic bone regeneration is a challenging process which involves the occurrence of sophisticated interactions. Although various polymeric scaffolds have been proposed for bone repair, research on osteoporotic bone regeneration remains practically limited. In particular, achieving satisfactory bone regeneration when using osteoporotic drugs is challenging including bisphosphonates. Here, a novel nitric oxide-releasing bioinspired scaffold with bioactive agents for the exquisite regeneration of osteoporotic bone is proposed. The bone-like biomimetic poly(lactic-co-glycolic acid) scaffold is first prepared in combination with organic/inorganic ECM and magnesium hydroxide as the base implant material. Nanoparticles containing bioactive agents of zinc oxide (ZO), alendronate, and BMP2 are incorporated to the biomimetic scaffold to impart multifunctionality such as anti-inflammation, angiogenesis, anti-osteoclastogenesis, and bone regeneration. Especially, nitric oxide (NO) generated from ZO stimulates the activity of cGMP and protein kinase G; in addition, ZO downregulates the RANKL/osteoprotegerin ratio by suppressing the Wnt/β-catenin signaling pathway. The new bone is formed much better in the osteoporotic rat model than in the normal model through the regulation of bone homeostasis via the scaffold. These synergistic effects suggest that such a bioinspired scaffold could be a comprehensive way to regenerate exceptionally osteoporotic bones.
Collapse
Affiliation(s)
- Jun‐Kyu Lee
- Department of Biomedical ScienceCHA University335 Pangyo‐ro, Bundang‐gu, Seongnam‐siGyeonggi‐do13488Republic of Korea
| | - Da‐Seul Kim
- Department of Biomedical ScienceCHA University335 Pangyo‐ro, Bundang‐gu, Seongnam‐siGyeonggi‐do13488Republic of Korea
- School of Integrative EngineeringChung‐Ang University84 Heukseok‐ro, Dongjak‐guSeoul06974Republic of Korea
| | - So‐Yeon Park
- Department of Biomedical ScienceCHA University335 Pangyo‐ro, Bundang‐gu, Seongnam‐siGyeonggi‐do13488Republic of Korea
- Division of BiotechnologyCollege of Life Sciences and BiotechnologyKorea UniversitySeongbuk‐guSeoul02841Republic of Korea
| | - Seung‐Woon Baek
- Department of Biomedical ScienceCHA University335 Pangyo‐ro, Bundang‐gu, Seongnam‐siGyeonggi‐do13488Republic of Korea
- Department of Biomedical EngineeringSKKU Institute for ConvergenceSungkyunkwan University (SKKU)2066 Seobu‐ro, Jangan‐gu, Suwon‐siGyeonggi‐do16419Republic of Korea
- Department of Intelligent Precision Healthcare ConvergenceSKKU Institute for ConvergenceSungkyunkwan University (SKKU)2066 Seobu‐ro, Jangan‐gu, Suwon‐siGyeonggi‐do16419Republic of Korea
| | - Ji‐Won Jung
- Department of Biomedical ScienceCHA University335 Pangyo‐ro, Bundang‐gu, Seongnam‐siGyeonggi‐do13488Republic of Korea
| | - Tae‐Hyung Kim
- School of Integrative EngineeringChung‐Ang University84 Heukseok‐ro, Dongjak‐guSeoul06974Republic of Korea
| | - Dong Keun Han
- Department of Biomedical ScienceCHA University335 Pangyo‐ro, Bundang‐gu, Seongnam‐siGyeonggi‐do13488Republic of Korea
| |
Collapse
|
13
|
Baek SW, Kim DS, Song DH, Kim HB, Lee S, Kim JH, Lee JK, Hong YJ, Park CG, Han DK. Reduced restenosis and enhanced re-endothelialization of functional biodegradable vascular scaffolds by everolimus and magnesium hydroxide. Biomater Res 2022; 26:86. [PMID: 36544178 PMCID: PMC9768885 DOI: 10.1186/s40824-022-00334-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Coronary artery disease is a cardiovascular disease with a high mortality and mortality rate in modern society. Vascular stent insertion to restore blood flow is essential to treat this disease. A fully biodegradable vascular scaffold (BVS) is a vascular poly (L-lactic acid) (PLLA) stent that is receiving growing interest as this is biodegradable in the body and does not require secondary removal surgery. However, acidic byproducts composed of PLLA produced during the biodegradation of the BVS can induce an inflammatory response. Magnesium hydroxide, a basic inorganic particle, neutralizes the acidic byproducts of PLLA. METHODS: In this study, we investigated using a BVS coated with everolimus and surface-modified magnesium hydroxide that suppresses smooth muscle cell proliferation and protects endothelial cells, respectively. The various characteristics of the functional stent were evaluated using in vitro and in vivo analyses. RESULTS: The BVS was successfully prepared with evenly coated everolimus and surface-modified magnesium hydroxide. A neutral pH value was maintained by magnesium hydroxide during degradation, and everolimus was released for one month. The coated BVS effectively inhibited protein adsorption and platelet adhesion, demonstrating excellent blood compatibility. In vitro analysis showed that BVS protects endothelial cells with magnesium hydroxide and selectively inhibits smooth muscle cell proliferation via everolimus treatment. The functional BVS was inserted into porcine coronary arteries for 28 days, and the results demonstrated that the restenosis and inflammation greatly decreased and re-endothelialization was enhanced as compared to others. CONCLUSIONS This study provides new insights into the design of drug-incorporated BVS stent for coronary artery disease.
Collapse
Affiliation(s)
- Seung-Woon Baek
- grid.410886.30000 0004 0647 3511Department of Biomedical Science, CHA University, 335 Pangyo-Ro, Bundang-Gu, Seongnam-Si, Gyeonggi 13488 Korea ,grid.264381.a0000 0001 2181 989XDepartment of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi 16419 Korea ,grid.264381.a0000 0001 2181 989XDepartment of Intelligent Precision Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi 16419 Korea
| | - Da-Seul Kim
- grid.410886.30000 0004 0647 3511Department of Biomedical Science, CHA University, 335 Pangyo-Ro, Bundang-Gu, Seongnam-Si, Gyeonggi 13488 Korea ,grid.254224.70000 0001 0789 9563School of Integrative Engineering, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974 Korea
| | - Duck Hyun Song
- grid.410886.30000 0004 0647 3511Department of Biomedical Science, CHA University, 335 Pangyo-Ro, Bundang-Gu, Seongnam-Si, Gyeonggi 13488 Korea
| | - Han Byul Kim
- grid.412484.f0000 0001 0302 820XThe Cardiovascular Convergence Research Center of Chonnam, National University Hospital Designated By Korea Ministry of Health and Welfare, 42 Jebong-ro, Dong-gu, Gwangju, 61469 Korea
| | - Semi Lee
- grid.410886.30000 0004 0647 3511Department of Biomedical Science, CHA University, 335 Pangyo-Ro, Bundang-Gu, Seongnam-Si, Gyeonggi 13488 Korea
| | - Jun Hyuk Kim
- grid.410886.30000 0004 0647 3511Department of Biomedical Science, CHA University, 335 Pangyo-Ro, Bundang-Gu, Seongnam-Si, Gyeonggi 13488 Korea
| | - Jun-Kyu Lee
- grid.410886.30000 0004 0647 3511Department of Biomedical Science, CHA University, 335 Pangyo-Ro, Bundang-Gu, Seongnam-Si, Gyeonggi 13488 Korea
| | - Young Joon Hong
- grid.412484.f0000 0001 0302 820XDivision of Cardiology of Chonnam, Cardiovascular Convergence Research Center Nominated By Korea Ministry of Health and Welfare, National University Hospital, 42 Jebong-ro, Dong-gu, Gwangju, 61469 Korea
| | - Chun Gwon Park
- grid.264381.a0000 0001 2181 989XDepartment of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi 16419 Korea ,grid.264381.a0000 0001 2181 989XDepartment of Intelligent Precision Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi 16419 Korea
| | - Dong Keun Han
- grid.410886.30000 0004 0647 3511Department of Biomedical Science, CHA University, 335 Pangyo-Ro, Bundang-Gu, Seongnam-Si, Gyeonggi 13488 Korea
| |
Collapse
|
14
|
Baek SW, Kim DS, Song DH, Lee S, Lee JK, Park SY, Kim JH, Kim TH, Park CG, Han DK. PLLA Composites Combined with Delivery System of Bioactive Agents for Anti-Inflammation and Re-Endothelialization. Pharmaceutics 2022; 14:pharmaceutics14122661. [PMID: 36559156 PMCID: PMC9782680 DOI: 10.3390/pharmaceutics14122661] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/24/2022] [Accepted: 11/25/2022] [Indexed: 12/02/2022] Open
Abstract
The development of a biodegradable vascular scaffold (BVS) for the treatment of cardiovascular diseases (CVDs) still requires some improvement. Among them, re-endothelialization and anti-inflammation are clinically important to restore vascular function. In this study, we proposed a coating system to deliver hydrophilic bioactive agents to BVS using nanoemulsion and drop-casting methods. The poly(L-lactide) (PLLA) scaffold containing magnesium hydroxide (MH) was coated on the surface with bioactive molecules such as polydeoxyribonucleotide (PDRN), L-arginine (Arg, R), and mesenchymal stem cell-derived extracellular vesicles (EVs). PDRN upregulates the expression of VEGF as one of the A2A receptor agonists; and Arg, synthesized into nitric oxide by intracellular eNOS, induces endothelialization. In particular, EVs, which are composed of a lipid bilayer and transfer bioactive materials such as protein and nucleic acid, regulate homeostasis in blood vessels. Such a bioactive agent coating system and its PLLA composite suggest a new platform for the treatment of cardiovascular dysfunction.
Collapse
Affiliation(s)
- Seung-Woon Baek
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Republic of Korea
- Department of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon-si 16419, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon-si 16419, Republic of Korea
| | - Da-Seul Kim
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Republic of Korea
- School of Integrative Engineering, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Duck Hyun Song
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Republic of Korea
| | - Semi Lee
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Republic of Korea
| | - Jun-Kyu Lee
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Republic of Korea
| | - So-Yeon Park
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Republic of Korea
- Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Jun Hyuk Kim
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Republic of Korea
| | - Tae-Hyung Kim
- School of Integrative Engineering, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Chun Gwon Park
- Department of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon-si 16419, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon-si 16419, Republic of Korea
| | - Dong Keun Han
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Republic of Korea
- Correspondence:
| |
Collapse
|
15
|
Cho S, Choi H, Jeong H, Kwon SY, Roh EJ, Jeong KH, Baek I, Kim BJ, Lee SH, Han I, Cha JM. Preclinical Study of Human Bone Marrow-Derived Mesenchymal Stem Cells Using a 3-Dimensional Manufacturing Setting for Enhancing Spinal Fusion. Stem Cells Transl Med 2022; 11:1072-1088. [PMID: 36180050 PMCID: PMC9585955 DOI: 10.1093/stcltm/szac052] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 06/12/2022] [Indexed: 11/29/2022] Open
Abstract
Spinal fusion surgery is a surgical technique that connects one or more vertebrae at the same time to prevent movement between the vertebrae. Although synthetic bone substitutes or osteogenesis-inducing recombinant proteins were introduced to promote bone union, the rate of revision surgery is still high due to pseudarthrosis. To promote successful fusion after surgery, stem cells with or without biomaterials were introduced; however, conventional 2D-culture environments have resulted in a considerable loss of the innate therapeutic properties of stem cells. Therefore, we conducted a preclinical study applying 3D-spheroids of human bone marrow-dewrived mesenchymal stem cells (MSCs) to a mouse spinal fusion model. First, we built a large-scale manufacturing platform for MSC spheroids, which is applicable to good manufacturing practice (GMP). Comprehensive biomolecular examinations, which include liquid chromatography-mass spectrometry and bioinformatics could suggest a framework of quality control (QC) standards for the MSC spheroid product regarding the identity, purity, viability, and potency. In our animal study, the mass-produced and quality-controlled MSC spheroids, either undifferentiated or osteogenically differentiated were well-integrated into decorticated bone of the lumbar spine, and efficiently improved angiogenesis, bone regeneration, and mechanical stability with statistical significance compared to 2D-cultured MSCs. This study proposes a GMP-applicable bioprocessing platform and QC directions of MSC spheroids aiming for their clinical application in spinal fusion surgery as a new bone graft substitute.
Collapse
Affiliation(s)
- Sumin Cho
- Department of Mechatronics Engineering, College of Engineering, Incheon National University, Incheon, Republic of Korea.,3D Stem Cell Bioengineering Laboratory, Research Institute for Engineering and Technology, Incheon National University, Incheon, Republic of Korea
| | - Hyemin Choi
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Hyundoo Jeong
- Department of Mechatronics Engineering, College of Engineering, Incheon National University, Incheon, Republic of Korea
| | - Su Yeon Kwon
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Eun Ji Roh
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Kwang-Hun Jeong
- Department of Mechatronics Engineering, College of Engineering, Incheon National University, Incheon, Republic of Korea.,3D Stem Cell Bioengineering Laboratory, Research Institute for Engineering and Technology, Incheon National University, Incheon, Republic of Korea
| | - Inho Baek
- Department of Biomedical Technology, Dongguk University, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Byoung Ju Kim
- Department of Biomedical Technology, Dongguk University, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Soo-Hong Lee
- Department of Biomedical Technology, Dongguk University, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Inbo Han
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Jae Min Cha
- Department of Mechatronics Engineering, College of Engineering, Incheon National University, Incheon, Republic of Korea.,3D Stem Cell Bioengineering Laboratory, Research Institute for Engineering and Technology, Incheon National University, Incheon, Republic of Korea
| |
Collapse
|
16
|
Highly porous multiple-cell-laden collagen/hydroxyapatite scaffolds for bone tissue engineering. Int J Biol Macromol 2022; 222:1264-1276. [DOI: 10.1016/j.ijbiomac.2022.09.249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/16/2022] [Accepted: 09/27/2022] [Indexed: 11/15/2022]
|
17
|
Guo Y, Zhao H, Lu J, Xu H, Hu T, Wu D. Preoperative Lymphocyte to Monocyte Ratio as a Predictive Biomarker for Disease Severity and Spinal Fusion Failure in Lumbar Degenerative Diseases Patients Undergoing Lumbar Fusion. J Pain Res 2022; 15:2879-2891. [PMID: 36124035 PMCID: PMC9482412 DOI: 10.2147/jpr.s379453] [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: 06/28/2022] [Accepted: 09/06/2022] [Indexed: 11/23/2022] Open
Abstract
Objective This study was designed to determine whether lymphocyte to monocyte ratio (LMR) correlated with the intervertebral disc degeneration (IDD) severity and the postoperative spinal fusion rate in patients with lumbar disc disease. Methods 303 patients undergoing posterior lumbar decompression and fusion were retrospectively analyzed. An examination of the blood count was performed before surgery. The cumulative grade was calculated by summing the pfirrmann grades of all lumbar discs. Grouping was based on the 50th percentile of cumulative grade and spinal fusion. The relationship between LMR and IDD severity and spinal fusion was explored using correlation analyses and logistic regression models. The receiver operating characteristic (ROC) curve was performed to measure model discrimination, and Hosmer-Lemeshow (H-L) test was used to measure calibration. Meanwhile, the ROC curve evaluated the discrimination ability of LMR in predicting severe degeneration and fusion failure. Results LMR was significantly lower in the severe degeneration group (cumulative grade > 18) than in the mild to moderate degeneration group (cumulative grade ≤ 18). Furthermore, the LMR of the fusion group was significantly higher than that of the non-fusion group. The multivariate binary logistic models revealed that LMR was an independently influencing factor of the severe degeneration and fusion failure (OR: 0.793, 95% CI: 0.638–0.987, p = 0.038; OR: 0.371, 95% CI: 0.258–0.532, p < 0.001). The models showed excellent discrimination and calibration. The area under the curve (AUC) of severe degeneration and fusion failure identified by LMR were 0.635 and 0.643, respectively, and the corresponding cut-off values were 3.16 and 3.90. Conclusion LMR is significantly associated with the risk of severe disc degeneration and spinal fusion failure.
Collapse
Affiliation(s)
- Youfeng Guo
- Department of Spine Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, People's Republic of China
| | - Haihong Zhao
- Department of Spine Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, People's Republic of China
| | - Jiawei Lu
- Department of Spine Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, People's Republic of China
| | - Haowei Xu
- Department of Spine Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, People's Republic of China
| | - Tao Hu
- Department of Spine Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, People's Republic of China
| | - Desheng Wu
- Department of Spine Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, People's Republic of China
| |
Collapse
|
18
|
Enhanced Mechanical Properties and Anti-Inflammation of Poly(L-Lactic Acid) by Stereocomplexes of PLLA/PDLA and Surface-Modified Magnesium Hydroxide Nanoparticles. Polymers (Basel) 2022; 14:polym14183790. [PMID: 36145934 PMCID: PMC9504497 DOI: 10.3390/polym14183790] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/01/2022] [Accepted: 09/07/2022] [Indexed: 12/29/2022] Open
Abstract
Poly(L-lactic acid) (PLLA), as a biodegradable polymer, has attracted attention for use as a biomaterial. In order to apply PLLA as a cardiovascular stent, stronger mechanical properties and anti-inflammatory effects against acidic by-products are required. In this study, PLLA/PDLA stereocomplex microparticles (SC) were developed and surface-modified magnesium hydroxide (MH) nanoparticles with oligolactide were combined with these PLLA composites. The SC improved the mechanical properties of the PLLA composites through the formation of stereocomplex structures. The surface-modified MH nanoparticles showed enhanced mechanical properties due to the stereocomplex structures formed by PLLA chains and inhibited inflammatory responses by pH neutralization as a result of MH. Additionally, the MH nanoparticles containing PLLA composites had antibacterial effects and increased the viability of human vascular endothelial cells. This technology is expected to have great potential in the development of PLLA composite materials for the production of various medical devices, such as cardiovascular stents.
Collapse
|
19
|
Alejo AL, McDermott S, Khalil Y, Ball HC, Robinson GT, Solorzano E, Alejo AM, Douglas J, Samson TK, Young JW, Safadi FF. A Pre-clinical Standard Operating Procedure for Evaluating Orthobiologics in an In Vivo Rat Spinal Fusion Model. JOURNAL OF ORTHOPAEDICS AND SPORTS MEDICINE 2022; 4:224-240. [PMID: 36203492 PMCID: PMC9534599 DOI: 10.26502/josm.511500060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The rat animal model is a cost effective and reliable model used in spinal pre-clinical research. Complications from various surgical procedures in humans often arise that were based on these pre-clinical animal models. Therefore safe and efficacious pre-clinical animal models are needed to establish continuity into clinical trials. A Standard Operating Procedure (SOP) is a validated method that allows researchers to safely and carefully replicate previously successful surgical techniques. Thus, the aim of this study is to describe in detail the procedures involved in a common rat bilateral posterolateral intertransverse spinal fusion SOP used to test the efficacy and safety different orthobiologics using a collagen-soaked sponge as an orthobiologic carrier. Only two orthobiologics are currently FDA approved for spinal fusion surgery which include recombinant bone morphogenetic protein 2 (rhBMP-2), and I-FACTOR. While there are many additional orthobiologics currently being tested, one way to show their safety profile and gain FDA approval, is to use well established pre-clinical animal models. A preoperative, intraoperative, and postoperative surgical setup including specific anesthesia and euthanasia protocols are outlined. Furthermore, we describe different postoperative methods used to validate the spinal fusion SOP, which include μCT analysis, histopathology, biomechanical testing, and blood analysis. This SOP can help increase validity, transparency, efficacy, and reproducibly in future rat spinal fusion surgery procedures.
Collapse
Affiliation(s)
- Andrew L Alejo
- College of Medicine, Northeast Ohio Medical University (NEOMED), Rootstown, OH, USA
- Musculoskeletal Research Group, NEOMED, Rootstown, OH, USA
| | - Scott McDermott
- Roper St. Francis Physician Partners Orthopaedics, Summerville, SC, USA
| | - Yusuf Khalil
- College of Medicine, Northeast Ohio Medical University (NEOMED), Rootstown, OH, USA
- Musculoskeletal Research Group, NEOMED, Rootstown, OH, USA
| | - Hope C Ball
- Musculoskeletal Research Group, NEOMED, Rootstown, OH, USA
| | - Gabrielle T Robinson
- College of Graduate Studies, NEOMED, Rootstown, OH, USA
- Musculoskeletal Research Group, NEOMED, Rootstown, OH, USA
| | - Ernesto Solorzano
- College of Graduate Studies, NEOMED, Rootstown, OH, USA
- Musculoskeletal Research Group, NEOMED, Rootstown, OH, USA
| | - Amanda M Alejo
- College of Medicine, Northeast Ohio Medical University (NEOMED), Rootstown, OH, USA
- Musculoskeletal Research Group, NEOMED, Rootstown, OH, USA
| | - Jacob Douglas
- College of Medicine, Northeast Ohio Medical University (NEOMED), Rootstown, OH, USA
- Musculoskeletal Research Group, NEOMED, Rootstown, OH, USA
| | - Trinity K Samson
- College of Medicine, Northeast Ohio Medical University (NEOMED), Rootstown, OH, USA
- College of Graduate Studies, NEOMED, Rootstown, OH, USA
- Musculoskeletal Research Group, NEOMED, Rootstown, OH, USA
| | - Jesse W Young
- College of Medicine, Northeast Ohio Medical University (NEOMED), Rootstown, OH, USA
- College of Graduate Studies, NEOMED, Rootstown, OH, USA
- Musculoskeletal Research Group, NEOMED, Rootstown, OH, USA
| | - Fayez F Safadi
- College of Medicine, Northeast Ohio Medical University (NEOMED), Rootstown, OH, USA
- College of Graduate Studies, NEOMED, Rootstown, OH, USA
- Musculoskeletal Research Group, NEOMED, Rootstown, OH, USA
- Rebecca D. Considine Research Institute, Akron Children's Hospital, Akron, OH, USA
- GPN Therapeutics Inc., Redi Zone NEOMED, Rootstown, OH, USA
| |
Collapse
|
20
|
Wu H, Zhao C, Lin K, Wang X. Mussel-Inspired Polydopamine-Based Multilayered Coatings for Enhanced Bone Formation. Front Bioeng Biotechnol 2022; 10:952500. [PMID: 35875492 PMCID: PMC9301208 DOI: 10.3389/fbioe.2022.952500] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 06/17/2022] [Indexed: 12/03/2022] Open
Abstract
Repairing bone defects remains a challenge in clinical practice and the application of artificial scaffolds can enhance local bone formation, but the function of unmodified scaffolds is limited. Considering different application scenarios, the scaffolds should be multifunctionalized to meet specific demands. Inspired by the superior adhesive property of mussels, polydopamine (PDA) has attracted extensive attention due to its universal capacity to assemble on all biomaterials and promote further adsorption of multiple external components to form PDA-based multilayered coatings with multifunctional property, which can induce synergistic enhancement of new bone formation, such as immunomodulation, angiogenesis, antibiosis and antitumor property. This review will summarize mussel-inspired PDA-based multilayered coatings for enhanced bone formation, including formation mechanism and biofunction of PDA coating, as well as different functional components. The synergistic enhancement of multiple functions for better bone formation will also be discussed. This review will inspire the design and fabrication of PDA-based multilayered coatings for different application scenarios and promote deeper understanding of their effect on bone formation, but more efforts should be made to achieve clinical translation. On this basis, we present a critical conclusion, and forecast the prospects of PDA-based multilayered coatings for bone regeneration.
Collapse
Affiliation(s)
| | | | - Kaili Lin
- *Correspondence: Kaili Lin, ; Xudong Wang,
| | | |
Collapse
|
21
|
Kim DH, Kim DS, Ha HJ, Jung JW, Baek SW, Baek SH, Kim TH, Lee JC, Hwang E, Han DK. Fat Graft with Allograft Adipose Matrix and Magnesium Hydroxide-Incorporated PLGA Microspheres for Effective Soft Tissue Reconstruction. Tissue Eng Regen Med 2022; 19:553-563. [PMID: 35312988 PMCID: PMC9130390 DOI: 10.1007/s13770-021-00426-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 12/16/2021] [Accepted: 12/20/2021] [Indexed: 10/18/2022] Open
Abstract
BACKGROUND Autologous fat grafting is one of the most common procedures used in plastic surgery to correct soft tissue deficiency or depression deformity. However, its clinical outcomes are often suboptimal, and lack of metabolic and architectural support at recipient sites affect fat survival leading to complications such as cyst formation, calcification. Extracellular matrix-based scaffolds, such as allograft adipose matrix (AAM) and poly(lactic-co-glycolic) acid (PLGA), have shown exceptional clinical promise as regenerative scaffolds. Magnesium hydroxide (MH), an alkaline ceramic, has attracted attention as a potential additive to improve biocompatibility. We attempted to combine fat graft with regenerative scaffolds and analyzed the changes and viability of injected fat graft in relation to the effects of injectable natural, and synthetic (PLGA/MH microsphere) biomaterials. METHODS In vitro cell cytotoxicity, angiogenesis of the scaffolds, and wound healing were evaluated using human dermal fibroblast cells. Subcutaneous soft-tissue integration of harvested fat tissue was investigated in vivo in nude mouse with random fat transfer protocol Fat integrity and angiogenesis were identified by qRT-PCR and immunohistochemistry. RESULTS In vitro cell cytotoxicity was not observed both in AAM and PLGA/MH with human dermal fibroblast. PLGA/MH and AAM showed excellent wound healing effect. In vivo, the AAM and PLGA/MH retained volume compared to that in the only fat group. And the PLGA/MH showed the highest angiogenesis and anti-inflammation. CONCLUSION In this study, a comparison of the volume retention effect and angiogenic ability between autologous fat grafting, injectable natural, and synthetic biomaterials will provide a reasonable basis for fat grafting.
Collapse
Affiliation(s)
- Dae-Hee Kim
- Department of Biomedical Engineering, College of Medicine, Seoul National University, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Da-Seul Kim
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam, 13488, Republic of Korea
- School of Integrative Engineering, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Hyun-Jeong Ha
- Department of Plastic and Reconstructive Surgery, CHA Bundang Medical Center, CHA University School of Medicine, 59 Yatap-ro, Bundang-gu, Seongnam, 13496, Republic of Korea
| | - Ji-Won Jung
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam, 13488, Republic of Korea
| | - Seung-Woon Baek
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam, 13488, Republic of Korea
| | - Seung Hwa Baek
- CHA Advanced Research Institute Center for Research & Development, Histological Analysis Team, 335 Pangyo-ro, Bundang-gu, Seongnam, 13488, Republic of Korea
| | - Tae-Hyung Kim
- School of Integrative Engineering, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Jung Chan Lee
- Department of Biomedical Engineering, College of Medicine, Seoul National University, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
| | - Euna Hwang
- Department of Plastic and Reconstructive Surgery, CHA Gangnam Medical Center, 566 Nonhyun-ro, Gangnam-gu, Seoul, 06135, Republic of Korea.
| | - Dong Keun Han
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam, 13488, Republic of Korea.
| |
Collapse
|
22
|
Zhao Y, Xing Y, Wang M, Huang Y, Xu H, Su Y, Zhao Y, Shang Y. Supramolecular Hydrogel Based on an Osteogenic Growth Peptide Promotes Bone Defect Repair. ACS OMEGA 2022; 7:11395-11404. [PMID: 35415354 PMCID: PMC8992256 DOI: 10.1021/acsomega.2c00501] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/16/2022] [Indexed: 05/13/2023]
Abstract
Current bone defect treatment strategies are associated with several risks and have major limitations. Therefore, it is necessary to develop an inexpensive growth factor delivery system that can be easily produced in large quantities and can promote long-term bone regeneration. An osteogenic growth peptide (OGP) is a 14 amino acid peptide with a short peptide sequence active fragment. In this study, we developed two OGP-based self-assembling supramolecular hydrogels (F- and G-sequence hydrogels) and investigated the in vitro and in vivo effects on proliferation and osteogenesis, including the mechanism of hydrogel-mediated bone defect repair. The hydrogels presented excellent biocompatibility and cell proliferation-promoting properties (1.5-1.7-fold increase). The hydrogels could effectively upregulate the expression of osteogenic factors, including RUNX2, BMP2, OCN, and OPN, to promote osteogenesis differentiation. Interestingly, 353 differentially expressed genes were identified in hBMSCs treated with hydrogels. The hydrogels were proved to be involved in the inflammatory pathways and folate-related pathways to mediate the osteogenesis differentiation. Furthermore, the therapeutic efficiency (bone volume/total volume, trabecular number, and bone mineral density) of hydrogels on bone regeneration in vivo was evaluated. The results showed that the hydrogels promoted bone formation in the early stage of bone defect healing. Taken together, this study was the first to develop and evaluate the properties of OGP-based self-assembling supramolecular hydrogels. Our study will provide inspiration for the development of delivering OGP for bone regeneration.
Collapse
Affiliation(s)
- Yanhong Zhao
- Hospital
of Stomatology, Tianjin Medical University, Tianjin 300070, People ’s Republic of China
| | - Yi Xing
- Hospital
of Stomatology, Tianjin Medical University, Tianjin 300070, People ’s Republic of China
| | - Min Wang
- Hospital
of Stomatology, Tianjin Medical University, Tianjin 300070, People ’s Republic of China
| | - Ying Huang
- Hospital
of Stomatology, Tianjin Medical University, Tianjin 300070, People ’s Republic of China
| | - Hainan Xu
- Hospital
of Stomatology, Tianjin Medical University, Tianjin 300070, People ’s Republic of China
| | - Yuran Su
- Hospital
of Stomatology, Tianjin Medical University, Tianjin 300070, People ’s Republic of China
| | - Yanmei Zhao
- Institute
of Disaster and Emergency Medicine, Tianjin
University, Tianjin 300072, People ’s Republic
of China
| | - Yuna Shang
- Tianjin
Key Laboratory of Structure and Performance for Functional Molecules,
College of Chemistry, Tianjin Normal University, Tianjin 300387, People ’s Republic of China
| |
Collapse
|
23
|
Osteogenic Differentiation of Human Adipose-Derived Stem Cells Seeded on a Biomimetic Spongiosa-like Scaffold: Bone Morphogenetic Protein-2 Delivery by Overexpressing Fascia. Int J Mol Sci 2022; 23:ijms23052712. [PMID: 35269855 PMCID: PMC8911081 DOI: 10.3390/ijms23052712] [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: 01/10/2022] [Revised: 02/18/2022] [Accepted: 02/21/2022] [Indexed: 01/27/2023] Open
Abstract
Human adipose-derived stem cells (hADSCs) have the capacity for osteogenic differentiation and, in combination with suitable biomaterials and growth factors, the regeneration of bone defects. In order to differentiate hADSCs into the osteogenic lineage, bone morphogenetic proteins (BMPs) have been proven to be highly effective, especially when expressed locally by route of gene transfer, providing a constant stimulus over an extended period of time. However, the creation of genetically modified hADSCs is laborious and time-consuming, which hinders clinical translation of the approach. Instead, expedited single-surgery gene therapy strategies must be developed. Therefore, in an in vitro experiment, we evaluated a novel growth factor delivery system, comprising adenoviral BMP-2 transduced fascia tissue in terms of BMP-2 release kinetics and osteogenic effects, on hADSCs seeded on an innovative biomimetic spongiosa-like scaffold. As compared to direct BMP-2 transduction of hADSCs or addition of recombinant BMP-2, overexpressing fascia provided a more uniform, constant level of BMP-2 over 30 days. Despite considerably higher BMP-2 peak levels in the comparison groups, delivery by overexpressing fascia led to a strong osteogenic response of hADSCs. The use of BMP-2 transduced fascia in combination with hADSCs may evolve into an expedited single-surgery gene transfer approach to bone repair.
Collapse
|
24
|
Qi H, Liu Y, Wu L, Liu C, Ni S, Liu Q, Ni X, Sun Q. Mg-HA-C/C Composites Promote Osteogenic Differentiation and Repair Bone Defects Through Inhibiting miR-16. Front Bioeng Biotechnol 2022; 10:838842. [PMID: 35186909 PMCID: PMC8854763 DOI: 10.3389/fbioe.2022.838842] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 01/10/2022] [Indexed: 11/13/2022] Open
Abstract
The hydroxyapatite (HA) coating on carbon/carbon (C/C) is reasonable and feasible to obtain bone graft materials with appropriate mechanical and biological properties. However, improvement of the physical and chemical properties of HA-C/C composites to promote bone regeneration and healing remains a challenge. In our present study, the HA coatings on C/C with magnesium (Mg) (Mg-HA-C/C) composites were synthesized that Ca (NO3)2, Mg (NO3)2, and NH4H2PO4 were mixed and coatings were made by electromagnetic induction deposition’s heating. As determined with in vitro experiments, Mg-HA-C/C composites containing 10 and 20% Mg decreased miR-16 levels, increased cell viability, elevated the levels of osteogenesis-related genes, and promoted osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) seeded on their surfaces. In a rat model of skull defects, compared to the control group, at 4 and 12 weeks after the operation, the bone volume fraction (BV/TV) of Mg-HA-C/C composite group was increased by 8.439 ± 2.681% and 23.837 ± 7.845%, as well as the trabecular thickness (Tb.Th) was 56.247 ± 24.238 μm and 114.911 ± 34.015 μm more. These composites also increased the levels of ALP and RUNX2 in skull. The Mg-HA-C/C composite-enhanced bone regeneration and healing were blocked by in situ injection of an miR-16 mimic lentivirus vector. Thus, Mg-HA-C/C composites promote osteogenic differentiation and repair bone defects through inhibiting miR-16.
Collapse
Affiliation(s)
- Hong Qi
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Yang Liu
- Department of Orthopedics, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Lu Wu
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Chun Liu
- Second People’s Hospital of Changzhou, Nanjing Medical University, Changzhou, China
| | - Su Ni
- Second People’s Hospital of Changzhou, Nanjing Medical University, Changzhou, China
| | - Qizhan Liu
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xinye Ni
- Second People’s Hospital of Changzhou, Nanjing Medical University, Changzhou, China
- *Correspondence: Xinye Ni, ; Qiang Sun,
| | - Qiang Sun
- Department of Orthopedics, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- *Correspondence: Xinye Ni, ; Qiang Sun,
| |
Collapse
|
25
|
Heo Y, Shin SW, Kim DS, Lee S, Park SY, Baek SW, Lee JK, Kim JH, Han DK. Bioactive PCL microspheres with enhanced biocompatibility and collagen production for functional hyaluronic acid dermal fillers. Biomater Sci 2022; 10:947-959. [DOI: 10.1039/d1bm01846a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Polymeric microspheres containing magnesium hydroxide (MH) and a bioactive agent (BA), such as apocynin (APO) and astaxanthin (ATX), have been prepared as functional dermal fillers with enhanced physicochemical and biological performance.
Collapse
Affiliation(s)
- Yun Heo
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi 13488, Republic of Korea
| | - Sang-Woo Shin
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi 13488, Republic of Korea
| | - Da-Seul Kim
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi 13488, Republic of Korea
- School of Integrative Engineering, Chung-Ang University, Dongjak-gu, Seoul 06911, Republic of Korea
| | - Semi Lee
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi 13488, Republic of Korea
| | - So-Yeon Park
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi 13488, Republic of Korea
- Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Seung-Woon Baek
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi 13488, Republic of Korea
- Department of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Suwon-si, Gyeonggi 16419, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Suwon-si, Gyeonggi 16419, Republic of Korea
| | - Jun-Kyu Lee
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi 13488, Republic of Korea
| | - Jun Hyuk Kim
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi 13488, Republic of Korea
| | - Dong Keun Han
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi 13488, Republic of Korea
| |
Collapse
|
26
|
Zhang Y, Jiang Y, Zou D, Yuan B, Ke HZ, Li W. Therapeutics for enhancement of spinal fusion: A mini review. J Orthop Translat 2021; 31:73-79. [PMID: 34934624 DOI: 10.1016/j.jot.2021.11.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 10/29/2021] [Accepted: 11/01/2021] [Indexed: 10/19/2022] Open
Abstract
Objective With the advances in biological technologies over the past 20 years, a number of new therapies to promote bone healing have been introduced. Particularly in the spinal surgery field, more unprecedented biological therapeutics become available to enhance spinal fusion success rate along with advanced instrumentation approaches. Yet surgeons may not have been well informed about their safety and efficacy profiles in order to improve clinical practices. Therefore there is a need to summarize the evidence and bring the latest progress to surgeons for better clinical services for patients. Methods We comprehensively reviewed the literatures in regard to the biological therapeutics for enhancement of spinal fusion published in the last two decades. Results Autograft bone is still the gold standard for bone grafting in spinal fusion surgery due to its good osteoconductive, osteoinductive, and osteogenic abilities. Accumulating evidence suggests that adding rhBMPs in combination with autograft effectively promotes the fusion rate and improves surgical outcomes. However, the stimulating effect on spinal fusion of other growth factors, including PDGF, VEGF, TGF-beta, and FGF, is not convincing, while Nell-1 and activin A exhibited preliminary efficacy. In terms of systemic therapeutic approaches, the osteoporosis drug Teriparatide has played a positive role in promoting bone healing after spinal surgery, while new medications such as denosumab and sclerostin antibodies still need further validation. Currently, other treatment, such as controlled-release formulations and carriers, are being studied for better releasing profile and the administration convenience of the active ingredients. Conclusion As the world's population continues to grow older, the number of spinal fusion cases grows substantially due to increased surgical needs for spinal degenerative disease (SDD). Critical advancements in biological therapeutics that promote spinal fusion have brought better clinical outcomes to patients lately. With the accumulation of higher-level evidence, the safety and efficacy of present and emerging products are becoming more evident. These emerging therapeutics will shift the landscape of perioperative therapy for the enhancement of spinal fusion.
Collapse
Affiliation(s)
- Yidan Zhang
- Angitia Biopharmaceuticals, Guangzhou, China
| | - Yu Jiang
- Orthopaedic Department, Peking University Third Hospital, Beijing, China.,Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Peking University Third Hospital, Beijing, China.,Beijing Key Laboratory of Spinal Disease Research, Peking University Third Hospital, Beijing, China
| | - Da Zou
- Orthopaedic Department, Peking University Third Hospital, Beijing, China.,Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Peking University Third Hospital, Beijing, China.,Beijing Key Laboratory of Spinal Disease Research, Peking University Third Hospital, Beijing, China
| | - Baozhi Yuan
- Angitia Biopharmaceuticals, Guangzhou, China
| | - Hua Zhu Ke
- Angitia Biopharmaceuticals, Guangzhou, China
| | - Weishi Li
- Orthopaedic Department, Peking University Third Hospital, Beijing, China.,Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Peking University Third Hospital, Beijing, China.,Beijing Key Laboratory of Spinal Disease Research, Peking University Third Hospital, Beijing, China
| |
Collapse
|
27
|
Lu X, Guo H, Li J, Sun T, Xiong M. Recombinant Human Bone Morphogenic Protein-2 Immobilized Fabrication of Magnesium Functionalized Injectable Hydrogels for Controlled-Delivery and Osteogenic Differentiation of Rat Bone Marrow-Derived Mesenchymal Stem Cells in Femoral Head Necrosis Repair. Front Cell Dev Biol 2021; 9:723789. [PMID: 34900987 PMCID: PMC8656218 DOI: 10.3389/fcell.2021.723789] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 08/23/2021] [Indexed: 11/13/2022] Open
Abstract
Femoral head necrosis (FHN) is a clinically progressive disease that leads to overwhelming complications without an effective therapeutic approach. In recent decades, transplantation of mesenchymal stem cells (MSCs) has played a promising role in the treatment of FHN in the initial stage; however, the success rate is still low because of unsuitable cell carriers and abridged osteogenic differentiation of the transplanted MSCs. Biopolymeric-derived hydrogels have been extensively applied as effective cell carriers and drug vesicles; they provide the most promising contributions in the fields of tissue engineering and regenerative medicine. However, the clinical potential of hydrogels may be limited because of inappropriate gelation, swelling, mechanical characteristics, toxicity in the cross-linking process, and self-healing ability. Naturally, gelated commercial hydrogels are not suitable for cell injection and infiltration because of their static network structure. In this study, we designed a novel thermogelling injectable hydrogel using natural silk fibroin-blended chitosan (CS) incorporated with magnesium (Mg) substitutes to improve physical cross-linking, stability, and cell osteogenic compatibility. The presented observations demonstrate that the developed injectable hydrogels can facilitate the controlled delivery of immobilized recombinant human bone morphogenic protein-2 (rhBMP-2) and rat bone marrow-derived MSCs (rBMSCs) with greater cell encapsulation efficiency, compatibility, and osteogenic differentiation. In addition, outcomes of in vivo animal studies established promising osteoinductive, bone mineral density, and bone formation rate after implantation of the injectable hydrogel scaffolds. Therefore, the developed hydrogels have great potential for clinical applications of FHN therapy.
Collapse
Affiliation(s)
- Xueliang Lu
- Department of Orthopedics, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
| | - Hongyu Guo
- Clinical Medical College, Henan University of Science and Technology, Luoyang, China
| | - Jiaju Li
- Clinical Medical College, Henan University of Science and Technology, Luoyang, China
| | - Tianyu Sun
- Clinical Medical College, Henan University of Science and Technology, Luoyang, China
| | - Mingyue Xiong
- Department of Orthopedics, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
| |
Collapse
|
28
|
Kim DS, Lee JK, Kim JH, Lee J, Kim DS, An S, Park SB, Kim TH, Rim JS, Lee S, Han DK. Advanced PLGA hybrid scaffold with a bioactive PDRN/BMP2 nanocomplex for angiogenesis and bone regeneration using human fetal MSCs. SCIENCE ADVANCES 2021; 7:eabj1083. [PMID: 34878837 PMCID: PMC8654289 DOI: 10.1126/sciadv.abj1083] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 10/16/2021] [Indexed: 05/14/2023]
Abstract
Biodegradable polymers have been used with various systems for tissue engineering. Among them, poly(lactic-co-glycolic) acid (PLGA) has been widely used as a biomaterial for bone regeneration because of its great biocompatibility and biodegradability properties. However, there remain substantial cruxes that the by-products of PLGA result in an acidic environment at the implanting site, and the polymer has a weak mechanical property. In our previous study, magnesium hydroxide (MH) and bone extracellular matrix are combined with a PLGA scaffold (PME) to improve anti-inflammation and mechanical properties and osteoconductivity. In the present study, the development of a bioactive nanocomplex (NC) formed along with polydeoxyribonucleotide and bone morphogenetic protein 2 (BMP2) provides synergistic abilities in angiogenesis and bone regeneration. This PME hybrid scaffold immobilized with NC (PME/NC) achieves outstanding performance in anti-inflammation, angiogenesis, and osteogenesis. Such an advanced PME/NC scaffold suggests an integrated bone graft substitute for bone regeneration.
Collapse
Affiliation(s)
- Da-Seul Kim
- Department of Biomedical Science, CHA University, Gyeonggi-do 13488, Republic of Korea
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Jun-Kyu Lee
- Department of Biomedical Science, CHA University, Gyeonggi-do 13488, Republic of Korea
| | - Jun Hyuk Kim
- Department of Biomedical Science, CHA University, Gyeonggi-do 13488, Republic of Korea
| | - Jaemin Lee
- Department of Biomedical Science, CHA University, Gyeonggi-do 13488, Republic of Korea
| | - Dong Seon Kim
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Sanghyun An
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Sung-Bin Park
- Department of Biomedical Science, CHA University, Gyeonggi-do 13488, Republic of Korea
| | - Tae-Hyung Kim
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Jong Seop Rim
- Fetal Stem Cell Research Center, CHA Advanced Research Institute, Gyeonggi-do 13488, Republic of Korea
| | - Soonchul Lee
- Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University, Gyeonggi-do 13496, Republic of Korea
| | - Dong Keun Han
- Department of Biomedical Science, CHA University, Gyeonggi-do 13488, Republic of Korea
| |
Collapse
|
29
|
Kang Y, Liu C, Wang M, Wang C, Yan YG, Wang WJ. A novel rat model of interbody fusion based on anterior lumbar corpectomy and fusion (ALCF). BMC Musculoskelet Disord 2021; 22:965. [PMID: 34794408 PMCID: PMC8603486 DOI: 10.1186/s12891-021-04822-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 10/26/2021] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Rats have been widely used as experimental animals when performing fundamental research because they are economical, rapidly reproducing, and heal quickly. While the rat interbody fusion model has been applied in basic studies, existing rat models generally have shortcomings, such as insufficiently simulating clinical surgery. The purpose of this study was to develop a novel rat model of interbody fusion which more closely represents clinical surgery. METHODS The internal fixation was designed based on physical measurements of the rats' lumbar spine. Then, ten rats divided into two groups (A and B) underwent anterior lumbar corpectomy and fusion of the L5 vertebrae. Groups A and B were sacrificed four and 8 weeks post-surgery, respectively. Micro-CT and histological examination were used to evaluate the model. Fusion rate, bone volume fraction (BV/TV), trabecular bone number (Tb.N), trabecular bone thickness (Tb.Th), and the area ratio of newly formed bone (NB) were calculated for quantitative analysis. RESULTS Based on the L5 body dimensions of individual rats, 3D-printed titanium cage of the appropriate size were printed. The operations were successfully completed in all ten rats, and X-ray confirmed that internal fixation was good without migration. Micro-CT suggested that fusion rates in group B (100%) were greater than group A (40%, P < 0.05). The BV/TV (B: 42.20 ± 10.50 vs. A: 29.02 ± 3.25, P < 0.05) and Tb.N (B: 4.66 ± 1.23 vs. A: 1.97 ± 0.40, P < 0.05) were greater in group B than A, and the Tb.Th in group B was lower than group A (B: 0.10 ± 0.04 vs. A: 0.15 ± 0.02, P < 0.05). Histomorphometry results demonstrated that the area ratio of NB in group B were greater than group A (B: 35.72 ± 12.80 vs. A: 12.36 ± 16.93, P < 0.05). CONCLUSION A rat interbody fusion model based on anterior lumbar corpectomy and fusion has successfully been constructed and verified. It could provide a new choice for fundamental research using animal models of spinal fusion.
Collapse
Affiliation(s)
- Yu Kang
- The First Affiliated Hospital, Department of Spine Surgery, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- Department of Orthopedicsity, The First Affiliated Hospital of Anhui Medical University, Hefei, 230000, Anhui, China
| | - Chao Liu
- The First Affiliated Hospital, Department of Spine Surgery, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Ming Wang
- The First Affiliated Hospital, Department of Spine Surgery, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Cheng Wang
- The First Affiliated Hospital, Department of Spine Surgery, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| | - Yi-Guo Yan
- The First Affiliated Hospital, Department of Spine Surgery, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| | - Wen-Jun Wang
- The First Affiliated Hospital, Department of Spine Surgery, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
30
|
Baneshi N, Moghadas BK, Adetunla A, Yusof MYPM, Dehghani M, Khandan A, Saber-Samandari S, Toghraie D. Investigation the mechanical properties of a novel multicomponent scaffold coated with a new bio-nanocomposite for bone tissue engineering: Fabrication, simulation and characterization. JOURNAL OF MATERIALS RESEARCH AND TECHNOLOGY 2021; 15:5526-5539. [DOI: 10.1016/j.jmrt.2021.10.107] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
31
|
Comparison of Surface Functionalization of PLGA Composite to Immobilize Extracellular Vesicles. Polymers (Basel) 2021; 13:polym13213643. [PMID: 34771200 PMCID: PMC8587822 DOI: 10.3390/polym13213643] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/14/2021] [Accepted: 10/19/2021] [Indexed: 02/06/2023] Open
Abstract
Endothelialization by materials provides a promising approach for the rapid re-endothelialization of a cardiovascular implantation. Although previous studies have focused on improving endothelialization through the immobilization of bioactive molecules onto the surface of biodegradable implants, comparative studies of effective surface modification have not yet been reported. Here, we conducted a comparative study on the surface modification of poly(lactide-co-glycolide) (PLGA)-based composites to graft mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) using three different materials, fibronectin (FN), polyethylenimine (PEI), and polydopamine (PDA), which have different bond strengths of ligand–receptor interaction, ionic bond, and covalent bond, respectively. Further in vitro analysis exhibited that MSC-EVs released from all modified films sustainably, but the MSC-EVs grafted onto the surface coated with PEI are more effective than other groups in increasing angiogenesis and reducing the inflammatory responses in endothelial cells. Therefore, the overall results demonstrated that PEI is a desirable coating reagent for the immobilization of MSC-EVs on the surface of biodegradable implants.
Collapse
|
32
|
Baek SW, Song DH, Lee HI, Kim DS, Heo Y, Kim JH, Park CG, Han DK. Poly(L-Lactic Acid) Composite with Surface-Modified Magnesium Hydroxide Nanoparticles by Biodegradable Oligomer for Augmented Mechanical and Biological Properties. MATERIALS (BASEL, SWITZERLAND) 2021; 14:5869. [PMID: 34640265 PMCID: PMC8510474 DOI: 10.3390/ma14195869] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/29/2021] [Accepted: 10/05/2021] [Indexed: 12/31/2022]
Abstract
Poly(L-lactic acid) (PLLA) has attracted a great deal of attention for its use in biomedical materials such as biodegradable vascular scaffolds due to its high biocompatibility. However, its inherent brittleness and inflammatory responses by acidic by-products of PLLA limit its application in biomedical materials. Magnesium hydroxide (MH) has drawn attention as a potential additive since it has a neutralizing effect. Despite the advantages of MH, the MH can be easily agglomerated, resulting in poor dispersion in the polymer matrix. To overcome this problem, oligo-L-lactide-ε-caprolactone (OLCL) as a flexible character was grafted onto the surface of MH nanoparticles due to its acid-neutralizing effect and was added to the PLLA to obtain PLLA/MH composites. The pH neutralization effect of MH was maintained after surface modification. In an in vitro cell experiment, the PLLA/MH composites including OLCL-grafted MH exhibited lower platelet adhesion, cytotoxicity, and inflammatory responses better than those of the control group. Taken together, these results prove that PLLA/MH composites including OLCL-grafted MH show excellent augmented mechanical and biological properties. This technology can be applied to biomedical materials for vascular devices such as biodegradable vascular scaffolds.
Collapse
Affiliation(s)
- Seung-Woon Baek
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Korea; (S.-W.B.); (D.H.S.); (H.I.L.); (D.-S.K.); (Y.H.); (J.H.K.)
- Department of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon-si 16419, Korea;
- Department of Intelligent Precision Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon-si 16419, Korea
| | - Duck Hyun Song
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Korea; (S.-W.B.); (D.H.S.); (H.I.L.); (D.-S.K.); (Y.H.); (J.H.K.)
| | - Ho In Lee
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Korea; (S.-W.B.); (D.H.S.); (H.I.L.); (D.-S.K.); (Y.H.); (J.H.K.)
| | - Da-Seul Kim
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Korea; (S.-W.B.); (D.H.S.); (H.I.L.); (D.-S.K.); (Y.H.); (J.H.K.)
- School of Integrative Engineering, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Korea
| | - Yun Heo
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Korea; (S.-W.B.); (D.H.S.); (H.I.L.); (D.-S.K.); (Y.H.); (J.H.K.)
| | - Jun Hyuk Kim
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Korea; (S.-W.B.); (D.H.S.); (H.I.L.); (D.-S.K.); (Y.H.); (J.H.K.)
| | - Chun Gwon Park
- Department of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon-si 16419, Korea;
- Department of Intelligent Precision Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon-si 16419, Korea
| | - Dong Keun Han
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Korea; (S.-W.B.); (D.H.S.); (H.I.L.); (D.-S.K.); (Y.H.); (J.H.K.)
| |
Collapse
|
33
|
The rough inhalable ciprofloxacin hydrochloride microparticles based on silk fibroin for non-cystic fibrosis bronchiectasis therapy with good biocompatibility. Int J Pharm 2021; 607:120974. [PMID: 34358540 DOI: 10.1016/j.ijpharm.2021.120974] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/13/2021] [Accepted: 08/01/2021] [Indexed: 01/03/2023]
Abstract
Non-cystic fibrosis bronchiectasis (NCFB) is a chronic respiratory disease, and the thick and viscous mucus covering on respiratory epithelia can entrap the inhaled drugs, resulting in compromised therapeutic efficiency. In order to solve this problem, the inhalable ciprofloxacin hydrochloride microparticles (CMs) based on silk fibroin (SF) and mannitol (MAN) were designed and developed. SF was applied to increase the loading efficiency of ciprofloxacin hydrochloride by strong electrostatic interactions. MAN could facilitate the penetration of drugs through mucus, which ensured the drugs could reach their targets before clearance. Furthermore, the aerodynamic performance of the inhalable microparticles could be tuned by changing the surface roughness to achieve a high fine particle fraction value (45.04%). The antibacterial effects of CMs were also confirmed by measuring the minimum inhibitory concentration against four different bacteria strains. Moreover, a series of experiments both in vitro and in vivo showed that CMs would not affect the lung function and induce the secretion of inflammatory cytokines in lungs, demonstrating their excellent biocompatibility and biosafety. Therefore, CMs might be a promising pulmonary drug delivery system for the treatment of NCFB.
Collapse
|
34
|
Rosehip Extract-Functionalized Magnesium Hydroxide Nanoparticles and Its Effect on Osteoblastic and Osteoclastic Cells. MATERIALS 2021; 14:ma14154172. [PMID: 34361365 PMCID: PMC8348532 DOI: 10.3390/ma14154172] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/19/2021] [Accepted: 07/24/2021] [Indexed: 01/21/2023]
Abstract
Considering the role of magnesium in bone metabolism and the increasing relevance of plant-mediated green-synthesis, this work compares the bone cytocompatibility of magnesium hydroxide nanoparticles (NPs) produced by using pure water, Mg(OH)2, or a rosehip (RH) aqueous extract, Mg(OH)2RH. The NPs were evaluated for dose- and time-dependent effects on human osteoblastic and osteoclastic response, due to the direct involvement of the two cell types in bone metabolism. Mg(OH)2 NPs presented nanoplatelet-like morphology (mean diameter ~90 nm) and a crystalline structure (XRD analysis); the RH-mediated synthesis yielded smaller rounded particles (mean diameter <10 nm) with decreased crystallinity. On the ATR-FTIR spectra, both NPs presented the characteristic Mg-OH peaks; Mg(OH)2RH exhibited additional vibration bands associated with the presence of phytochemicals. On osteoblastic cells, NPs did not affect cell growth and morphology but significantly increased alkaline phosphatase (ALP) activity; on osteoclastic cells, particles had little effect in protein content, tartrate-resistant acid phosphatase (TRAP) activity, percentage of multinucleated cells, and cell area. However, compared with Mg(OH)2, Mg(OH)2RH increased osteoblastic differentiation by inducing ALP activity and promoting the expression of Runx2, SP7, Col1a1, and ALP, and had a negative effect on the expression of the osteoclastic genes NFATC1, CA2, and CTSK. These observations suggest the potential usefulness of Mg(OH)2RH NPs in bone regeneration.
Collapse
|
35
|
Promotion of Bone Regeneration Using Bioinspired PLGA/MH/ECM Scaffold Combined with Bioactive PDRN. MATERIALS 2021; 14:ma14154149. [PMID: 34361342 PMCID: PMC8348682 DOI: 10.3390/ma14154149] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/26/2021] [Accepted: 07/22/2021] [Indexed: 12/16/2022]
Abstract
Current approaches of biomaterials for the repair of critical-sized bone defects still require immense effort to overcome numerous obstacles. The biodegradable polymer-based scaffolds have been required to expand further function for bone tissue engineering. Poly(lactic-co-glycolic) acid (PLGA) is one of the most common biopolymers owing to its biodegradability for tissue regenerations. However, there are major clinical challenges that the byproducts of the PLGA cause an acidic environment of implanting site. The critical processes in bone repair are osteogenesis, angiogenesis, and inhibition of excessive osteoclastogenesis. In this study, the porous PLGA (P) scaffold was combined with magnesium hydroxide (MH, M) and bone-extracellular matrix (bECM, E) to improve anti-inflammatory ability and osteoconductivity. Additionally, the bioactive polydeoxyribonucleotide (PDRN, P) was additionally incorporated in the existing PME scaffold. The prepared PMEP scaffold has pro-osteogenic and pro-angiogenic effects and inhibition of osteoclast due to the PDRN, which interacts with the adenosine A2A receptor agonist that up-regulates expression of vascular endothelial growth factor (VEGF) and down-regulates inflammatory cytokines. The PMEP scaffold has superior biological properties for human bone-marrow mesenchymal stem cells (hBMSCs) adhesion, proliferation, and osteogenic differentiation in vitro. Moreover, the gene expressions related to osteogenesis and angiogenesis of hBMSCs increased and the inflammatory factors decreased on the PMEP scaffold. In conclusion, it provides a promising strategy and clinical potential candidate for bone tissue regeneration and repairing bone defects.
Collapse
|
36
|
Lee HI, Heo Y, Baek SW, Kim DS, Song DH, Han DK. Multifunctional Biodegradable Vascular PLLA Scaffold with Improved X-ray Opacity, Anti-Inflammation, and Re-Endothelization. Polymers (Basel) 2021; 13:polym13121979. [PMID: 34208677 PMCID: PMC8234203 DOI: 10.3390/polym13121979] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 06/14/2021] [Accepted: 06/15/2021] [Indexed: 11/16/2022] Open
Abstract
Poly(L-lactic acid) (PLLA) has been used as a biodegradable vascular scaffold (BVS) material due to high mechanical property, biodegradability, and biocompatibility. However, acidic byproducts from hydrolysis of PLLA reduce the pH after the surrounding implanted area and cause inflammatory responses. As a result, severe inflammation, thrombosis, and in-stent restenosis can occur after implantation by using BVS. Additionally, polymers such as PLLA could not find on X-ray computed tomography (CT) because of low radiopacity. To this end, here, we fabricated PLLA films as the surface of BVS and divided PLLA films into two coating layers. At the first layer, PLLA film was coated by 2,3,5-triiodobenzoic acid (TIBA) and magnesium hydroxide (MH) with poly(D,L-lactic acid) (PDLLA) for radiopaque and neutralization of acidic environment, respectively. The second layer of coated PLLA films is composed of polydopamine (PDA) and then cystamine (Cys) for the generation of nitric oxide (NO) release, which is needed for suppression of smooth muscle cells (SMCs) and proliferation of endothelial cells (ECs). The characterization of the film surface was conducted via various analyses. Through the surface modification of PLLA films, they have multifunctional abilities to overcome problems of BVS effectively such as X-ray penetrability, inflammation, thrombosis, and neointimal hyperplasia. These results suggest that the modification of biodegradable PLLA using TIBA, MH, PDA, and Cys will have important potential in implant applications.
Collapse
Affiliation(s)
- Ho In Lee
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Korea; (H.I.L.); (Y.H.); (S.-W.B.); (D.-S.K.); (D.H.S.)
| | - Yun Heo
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Korea; (H.I.L.); (Y.H.); (S.-W.B.); (D.-S.K.); (D.H.S.)
| | - Seung-Woon Baek
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Korea; (H.I.L.); (Y.H.); (S.-W.B.); (D.-S.K.); (D.H.S.)
- Department of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon-si 16419, Korea
- Department of Intelligent Precision Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), 2066 Seobu-ro, Jangan-gu, Suwon-si 16419, Korea
| | - Da-Seul Kim
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Korea; (H.I.L.); (Y.H.); (S.-W.B.); (D.-S.K.); (D.H.S.)
- School of Integrative Engineering, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Korea
| | - Duck Hyun Song
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Korea; (H.I.L.); (Y.H.); (S.-W.B.); (D.-S.K.); (D.H.S.)
| | - Dong Keun Han
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si 13488, Korea; (H.I.L.); (Y.H.); (S.-W.B.); (D.-S.K.); (D.H.S.)
- Correspondence:
| |
Collapse
|
37
|
PLGA Microspheres Containing Hydrophobically Modified Magnesium Hydroxide Particles for Acid Neutralization-Mediated Anti-Inflammation. Tissue Eng Regen Med 2021; 18:613-622. [PMID: 33877618 DOI: 10.1007/s13770-021-00338-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/03/2021] [Accepted: 03/10/2021] [Indexed: 10/21/2022] Open
Abstract
BACKGROUND Poly(lactic-co-glycolic acid) (PLGA) microspheres have been actively used in various pharmaceutical formulations because they can sustain active pharmaceutical ingredient release and are easy to administer into the body using a syringe. However, the acidic byproducts produced by the decomposition of PLGA cause inflammatory reactions in surrounding tissues, limiting biocompatibility. Magnesium hydroxide (MH), an alkaline ceramic, has attracted attention as a potential additive because it has an acid-neutralizing effect. METHODS To improve the encapsulation efficiency of hydrophilic MH, the MH particles were capped with hydrophobic ricinoleic acid (RA-MH). PLGA microspheres encapsulated with RA-MH particles were manufactured by the O/W method. To assess the in vitro cytotoxicity of the degradation products of PLGA, MH/PLGA, and RA-MH/PLGA microspheres, CCK-8 and Live/Dead assays were performed with NIH-3T3 cells treated with different concentrations of their degradation products. In vitro anti-inflammatory effect of RA-MH/PLGA microspheres was evaluated with quantitative measurement of pro-inflammatory cytokines. RESULTS The synthesized RA-MH was encapsulated in PLGA microspheres and displayed more than four times higher loading content than pristine MH. The PLGA microspheres encapsulated with RA-MH had an acid-neutralizing effect better than that of the control group. In an in vitro cell experiment, the degradation products obtained from RA-MH/PLGA microspheres exhibited higher biocompatibility than the degradation products obtained from PLGA microspheres. Additionally, the RA-MH/PLGA microsphere group showed an excellent anti-inflammatory effect. CONCLUSION Our results proved that RA-MH-encapsulated PLGA microspheres showed excellent biocompatibility with an anti-inflammatory effect. This technology can be applied to drug delivery and tissue engineering to treat various incurable diseases in the future.
Collapse
|
38
|
Hu Y, Zhao QW, Wang ZC, Fang QQ, Zhu H, Hong DS, Liang XG, Lou D, Tan WQ. Co-transfection with BMP2 and FGF2 via chitosan nanoparticles potentiates osteogenesis in human adipose-derived stromal cells in vitro. J Int Med Res 2021; 49:300060521997679. [PMID: 33769121 PMCID: PMC8166400 DOI: 10.1177/0300060521997679] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 02/04/2021] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE To investigate if co-transfection of human bone morphogenetic protein 2 (BMP-2, BMP2) and human fibroblast growth factor 2 (FGF2, FGF2) via chitosan nanoparticles promotes osteogenesis in human adipose tissue-derived stem cells (ADSCs) in vitro. MATERIALS AND METHODS Recombinant BMP2 and/or FGF2 expression vectors were constructed and packaged into chitosan nanoparticles. The chitosan nanoparticles were characterized by atomic force microscopy. Gene and protein expression levels of BMP-2 and FGF2 in ADSCs in vitro were evaluated by real-time polymerase chain reaction (PCR), western blot, and enzyme-linked immunosorbent assay. Osteocalcin (OCN) and bone sialoprotein (BSP) gene expression were also evaluated by real-time PCR to assess osteogenesis. RESULTS The prepared chitosan nanoparticles were spherical with a relatively homogenous size distribution. The BMP2 and FGF2 vectors were successfully transfected into ADSCs. BMP-2 and FGF2 mRNA and protein levels were significantly up-regulated in the co-transfection group compared with the control group. OCN and BSP mRNA levels were also significantly increased in the co-transfection group compared with cells transfected with BMP2 or FGF2 alone, suggesting that co-transfection significantly enhanced osteogenesis. CONCLUSIONS Co-transfection of human ADSCs with BMP2/FGF2 via chitosan nanoparticles efficiently promotes the osteogenic properties of ADSCs in vitro.
Collapse
Affiliation(s)
- Ying Hu
- Department of Plastic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Qing-Wei Zhao
- Department of Clinical Pharmacy, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Zheng-Cai Wang
- Department of Plastic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Qing-Qing Fang
- Department of Plastic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
| | - He Zhu
- Department of Clinical Pharmacy, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Dong-Sheng Hong
- Department of Clinical Pharmacy, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Xing-Guang Liang
- Department of Clinical Pharmacy, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Dong Lou
- Department of Plastic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Wei-Qiang Tan
- Department of Plastic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
| |
Collapse
|
39
|
The Effect of Selenium Nanoparticles on the Osteogenic Differentiation of MC3T3-E1 Cells. NANOMATERIALS 2021; 11:nano11020557. [PMID: 33672352 PMCID: PMC7926403 DOI: 10.3390/nano11020557] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 02/10/2021] [Accepted: 02/17/2021] [Indexed: 12/13/2022]
Abstract
Reactive oxygen species (ROS) regulate various functions of cells, including cell death, viability, and differentiation, and nanoparticles influence ROS depending on their size and shape. Selenium is known to regulate various physiological functions, such as cell differentiations and anti-inflammatory functions, and plays an important role in the regulation of ROS as an antioxidant. This study aims to investigate the effect of selenium nanoparticles (SeNPs) on the differentiation of osteogenic MC3T3-E1 cells. After fabrication of SeNPs with a size of 25.3 ± 2.6 nm, and confirmation of its oxidase-like activity, SeNPs were added to MC3T3-E1 cells with or without H2O2: 5~20 μg/mL SeNPs recovered cells damaged by 200 μM H2O2 via the intracellular ROS downregulating role of SeNPs, revealed by the ROS staining assay. The increase in osteogenic maturation with SeNPs was gradually investigated by expression of osteogenic genes at 3 and 7 days, Alkaline phosphatase activity staining at 14 days, and Alizarin red S staining at 28 days. Therefore, the role of SeNPs in regulating ROS and their therapeutic effects on the differentiation of MC3T3-E1 cells were determined, leading to possible applications for bone treatment.
Collapse
|
40
|
Bedair TM, Heo Y, Ryu J, Bedair HM, Park W, Han DK. Biocompatible and functional inorganic magnesium ceramic particles for biomedical applications. Biomater Sci 2021; 9:1903-1923. [PMID: 33506843 DOI: 10.1039/d0bm01934h] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Magnesium ceramics hold promise for numerous biological applications. This review covers the synthesis of magnesium ceramic particles with specific morphologies and potential modification techniques. Magnesium ceramic particles possess multiple characteristics directly applicable to human biology; they are anti-inflammatory, antibacterial, antiviral, and offer anti-cancer effects. Based on these advantages, magnesium hydroxide nanoparticles have been extensively utilized across biomedical fields. In a vascular stent, the incorporation of magnesium ceramic nanoparticles enhances re-endothelialization. Additionally, tissue regeneration for bone, cartilage, and kidney can be promoted by magnesium ceramics. This review enables researchers to identify the optimum synthetic conditions to prepare magnesium ceramics with specific morphologies and sizes and select the appropriate modification protocols. It is also intended to elucidate the desirable physicochemical properties and biological benefits of magnesium ceramics.
Collapse
Affiliation(s)
- Tarek M Bedair
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi 13488, Korea.
| | | | | | | | | | | |
Collapse
|