1
|
Yousefi F, Foster LA, Selim OA, Zhao C. Integrating Physical and Biochemical Cues for Muscle Engineering: Scaffolds and Graft Durability. Bioengineering (Basel) 2024; 11:1245. [PMID: 39768063 PMCID: PMC11673930 DOI: 10.3390/bioengineering11121245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/26/2024] [Accepted: 11/28/2024] [Indexed: 01/11/2025] Open
Abstract
Muscle stem cells (MuSCs) are essential for skeletal muscle regeneration, influenced by a complex interplay of mechanical, biochemical, and molecular cues. Properties of the extracellular matrix (ECM) such as stiffness and alignment guide stem cell fate through mechanosensitive pathways, where forces like shear stress translate into biochemical signals, affecting cell behavior. Aging introduces senescence which disrupts the MuSC niche, leading to reduced regenerative capacity via epigenetic alterations and metabolic shifts. Transplantation further challenges MuSC viability, often resulting in fibrosis driven by dysregulated fibro-adipogenic progenitors (FAPs). Addressing these issues, scaffold designs integrated with pharmacotherapy emulate ECM environments, providing cues that enhance graft functionality and endurance. These scaffolds facilitate the synergy between mechanotransduction and intracellular signaling, optimizing MuSC proliferation and differentiation. Innovations utilizing human pluripotent stem cell-derived myogenic progenitors and exosome-mediated delivery exploit bioactive properties for targeted repair. Additionally, 3D-printed and electrospun scaffolds with adjustable biomechanical traits tackle scalability in treating volumetric muscle loss. Advanced techniques like single-cell RNA sequencing and high-resolution imaging unravel muscle repair mechanisms, offering precise mapping of cellular interactions. Collectively, this interdisciplinary approach fortifies tissue graft durability and MuSC maintenance, propelling therapeutic strategies for muscle injuries and degenerative diseases.
Collapse
Affiliation(s)
- Farbod Yousefi
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (F.Y.); (L.A.F.); (O.A.S.)
| | - Lauren Ann Foster
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (F.Y.); (L.A.F.); (O.A.S.)
- Atlanta Veterans Affairs Medical Center, Emory University School of Medicine, Atlanta, GA 30307, USA
| | - Omar A. Selim
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (F.Y.); (L.A.F.); (O.A.S.)
| | - Chunfeng Zhao
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA; (F.Y.); (L.A.F.); (O.A.S.)
| |
Collapse
|
2
|
Haghwerdi F, Haririan I, Soleimani M. Chondrogenic potential of PMSCs cultured on chondroitin sulfate/gelatin-modified DBM scaffold. BIOIMPACTS : BI 2024; 15:30003. [PMID: 40161935 PMCID: PMC11954754 DOI: 10.34172/bi.2023.30003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 10/15/2023] [Accepted: 10/15/2023] [Indexed: 04/02/2025]
Abstract
Introduction Osteoarthritis is one of the most common orthopedic diseases that gradually causes wear and damage to the articular Subchondral bone due to the destruction of articular cartilage. One of the basic challenges in cartilage tissue engineering is the choice of scaffold. In the design of the cartilage scaffold, it is useful to consider parameters such as porosity, water absorption, high mechanical resistance, biocompatibility, and biodegradability. Therefore, in this study, demineralized bone matrix (DBM), which inherently has these characteristics to some extent, was chosen as the basic scaffold. Methods The gelatin/DBM (G/DBM) and the chondroitin sulfate-gelatin/DBM (GCS/DBM) scaffolds were prepared, respectively, by incorporating gelatin or chondroitin sulfate/gelatin solution inside DBM pores, freeze-drying and crosslinking with EDC/NHS. The physicochemical, biological characteristics and chondrogenic potential of scaffolds were studied. Results According to the SEM results, the size of the DBM pores in the G/DBM and GCS/DBM scaffolds decreased (from almost 100-1500 µm to less than 200 µm), which reduced cell escape compared to the DBM scaffold. Also, crosslinking the scaffolds has greatly increased their compressive E-modulus (more than 8 times). The cytocompatibility and non- toxicity of all scaffolds were confirmed by acridine orange/ethidium bromide (AO/EB) staining. The evaluation results of chondrogenic differentiation of placenta-derived mesenchymal stem cells (PMSCs) on modified scaffolds, using the real-time PCR method, showed that the presence of CS in the GCS/DBM scaffold improved the expression of chondrogenesis markers such as Aggrecan (AGC) (~4 times) and collagen 2 (COL-2) (~2.2 times) compared to the DBM scaffold. Also, Alcian blue staining and immunohistochemical analyses of the scaffolds showed denser and more coherent GAGs and COL-2 protein synthesis on the GCS/DBM than the G/DBM and DBM scaffolds. Conclusion According to the results, the GCS/DBM scaffold can be a suitable scaffold for cartilage tissue engineering.
Collapse
Affiliation(s)
- Fatemeh Haghwerdi
- Department of Pharmaceutical Biomaterials and Medical Biomaterial Research Center (MBRC), Faculty of Pharmacy, Tehran University of Medical Science, Tehran, Iran
| | - Ismaeil Haririan
- Department of Pharmaceutical Biomaterials and Medical Biomaterial Research Center (MBRC), Faculty of Pharmacy, Tehran University of Medical Science, Tehran, Iran
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoud Soleimani
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
3
|
Aslam Khan MU, Aslam MA, Bin Abdullah MF, Stojanović GM. Current Perspectives of Protein in Bone Tissue Engineering: Bone Structure, Ideal Scaffolds, Fabrication Techniques, Applications, Scopes, and Future Advances. ACS APPLIED BIO MATERIALS 2024; 7:5082-5106. [PMID: 39007509 DOI: 10.1021/acsabm.4c00362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
In view of their exceptional approach, excellent inherent biocompatibility and biodegradability properties, and interaction with the local extracellular matrix, protein-based polymers have received attention in bone tissue engineering, which is a multidisciplinary field that repairs and regenerates fractured bones. Bone is a multihierarchical complex structure, and it performs several essential biofunctions, including maintaining mineral balance and structural support and protecting soft organs. Protein-based polymers have gained interest in developing ideal scaffolds as emerging biomaterials for bone fractured healing and regeneration, and it is challenging to design ideal bone substitutes as perfect biomaterials. Several protein-based polymers, including collagen, keratin, gelatin, serum albumin, etc., are potential materials due to their inherent cytocompatibility, controlled biodegradability, high biofunctionalization, and tunable mechanical characteristics. While numerous studies have indicated the encouraging possibilities of proteins in BTE, there are still major challenges concerning their biodegradability, stability in physiological conditions, and continuous release of growth factors and bioactive molecules. Robust scaffolds derived from proteins can be used to replace broken or diseased bone with a biocompatible substitute; proteins, being biopolymers, provide excellent scaffolds for bone tissue engineering. Herein, recent developments in protein polymers for cutting-edge bone tissue engineering are addressed in this review within 3-5 years, with a focus on the significant challenges and future perspectives. The first section discusses the structural fundamentals of bone anatomy and ideal scaffolds, and the second section describes the fabrication techniques of scaffolds. The third section highlights the importance of proteins and their applications in BTE. Hence, the recent development of protein polymers for state-of-the-art bone tissue engineering has been discussed, highlighting the significant challenges and future perspectives.
Collapse
Affiliation(s)
- Muhammad Umar Aslam Khan
- Department of Mechanical and Industrial Engineering, Qatar University, Doha 2713, Qatar
- Biomedical Research Center, Qatar University, Doha 2713, Qatar
| | - Muhammad Azhar Aslam
- Department of Physics, University of Engineering and Technology, Lahore 39161, Pakistan
| | - Mohd Faizal Bin Abdullah
- Oral and Maxillofacial Surgery Unit, School of Dental Sciences, Universiti Sains Malaysia, Health Campus Kubang Kerian 16150, Kota Bharu, Kelantan, Malaysia
- Oral and Maxillofacial Surgery Unit, Hospital Universiti Sains Malaysia, Universiti Sains Malaysia, Health Campus Kubang Kerian 16150, Kota Bharu, Kelantan, Malaysia
| | - Goran M Stojanović
- Faculty of Technical Sciences, University of Novi Sad, T. D. Obradovica 6, 21000 Novi Sad, Serbia
| |
Collapse
|
4
|
Yin H, Wang L, Hur SJ, Liu Y, Cong P, Liu H, Jiang X, Zheng H, Xue C. Cell-Cultured Fish Meat via Scale-Up Expansion of Carassius auratus Skeletal Muscle Cells Using Edible Porous Microcarriers and Quality Evaluation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:16475-16483. [PMID: 38987705 DOI: 10.1021/acs.jafc.4c03586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
Emerging technologies for cell-cultured fish meat as an environmentally friendly protein source for humans still have many obstacles, including large-scale production of high-quality cells, differentiation and bioassembly of cellular material, and improvement of the quality of meat products. Here, we used edible porous microcarriers as scaffolds to support scalable skeletal muscle cell expansion to prepare centimeter-scale cell-cultured fish (CCM) of Carassius auratus for the first time. The quality of CCM was assessed by analyzing the texture, nutrition, flavor, and safety. The results indicated that CCM demonstrated a softer texture than natural fish due to a high moisture content. CCM contained higher protein and lower fat contents, with no significant difference in energy from natural golden crucian carp meat (NGM). CCM had better digestible properties, and 17 volatile components were identified in CCM, ten cocontained compared to NGM. ELISA quantified penicillin, streptomycin, vitamin D, and insulin residues as risk factors in CCM. In conclusion, we utilized edible porous microcarriers to scale-up the expansion of Carassius auratus skeletal muscle cells and bioassembled high-quality CCM of Carassius auratus for the first time, which represents a state-of-the-art protocol applicable to different fish species and even to other economic animals and provides a theoretical basis for scaling up cell-cultured meat production.
Collapse
Affiliation(s)
- Haowen Yin
- State Key Laboratory of Marine Food Processing and Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao 266003, P.R. China
- Qingdao Institute of Marine Bioresources for Nutrition & Health Innovation, Qingdao 266109, P.R. China
| | - Lei Wang
- State Key Laboratory of Marine Food Processing and Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao 266003, P.R. China
| | - Sun Jin Hur
- Department of Animal Science and Technology, Chung-Ang University, Anseong 17546, Republic of Korea
| | - Ye Liu
- Qingdao Institute of Marine Bioresources for Nutrition & Health Innovation, Qingdao 266109, P.R. China
| | - Peixu Cong
- State Key Laboratory of Marine Food Processing and Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao 266003, P.R. China
| | - Hongying Liu
- Qingdao Institute of Marine Bioresources for Nutrition & Health Innovation, Qingdao 266109, P.R. China
| | - Xiaoming Jiang
- State Key Laboratory of Marine Food Processing and Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao 266003, P.R. China
- Qingdao Institute of Marine Bioresources for Nutrition & Health Innovation, Qingdao 266109, P.R. China
| | - Hongwei Zheng
- State Key Laboratory of Marine Food Processing and Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao 266003, P.R. China
- Qingdao Institute of Marine Bioresources for Nutrition & Health Innovation, Qingdao 266109, P.R. China
| | - Changhu Xue
- State Key Laboratory of Marine Food Processing and Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao 266003, P.R. China
- Qingdao Institute of Marine Bioresources for Nutrition & Health Innovation, Qingdao 266109, P.R. China
| |
Collapse
|
5
|
Jiang M, Zhang GH, Yu Y, Zhao YH, Liu J, Zeng Q, Feng MY, Ye F, Xiong DS, Wang L, Zhang YN, Yu L, Wei JJ, He LB, Zhi W, Du XR, Li NJ, Han CL, Yan HQ, Zhou ZT, Miao YB, Wang W, Liu WX. De novo design of a nanoregulator for the dynamic restoration of ovarian tissue in cryopreservation and transplantation. J Nanobiotechnology 2024; 22:330. [PMID: 38862987 PMCID: PMC11167790 DOI: 10.1186/s12951-024-02602-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 05/28/2024] [Indexed: 06/13/2024] Open
Abstract
The cryopreservation and transplantation of ovarian tissue underscore its paramount importance in safeguarding reproductive capacity and ameliorating reproductive disorders. However, challenges persist in ovarian tissue cryopreservation and transplantation (OTC-T), including the risk of tissue damage and dysfunction. Consequently, there has been a compelling exploration into the realm of nanoregulators to refine and enhance these procedures. This review embarks on a meticulous examination of the intricate anatomical structure of the ovary and its microenvironment, thereby establishing a robust groundwork for the development of nanomodulators. It systematically categorizes nanoregulators and delves deeply into their functions and mechanisms, meticulously tailored for optimizing ovarian tissue cryopreservation and transplantation. Furthermore, the review imparts valuable insights into the practical applications and obstacles encountered in clinical settings associated with OTC-T. Moreover, the review advocates for the utilization of microbially derived nanomodulators as a potent therapeutic intervention in ovarian tissue cryopreservation. The progression of these approaches holds the promise of seamlessly integrating nanoregulators into OTC-T practices, thereby heralding a new era of expansive applications and auspicious prospects in this pivotal domain.
Collapse
Affiliation(s)
- Min Jiang
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China
| | - Guo-Hui Zhang
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China
| | - Yuan Yu
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Yu-Hong Zhao
- School of Clinical Laboratory Medicine, Chengdu Medical College, Chengdu, 610083, China
| | - Jun Liu
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Qin Zeng
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China
| | - Meng-Yue Feng
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Fei Ye
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China
| | - Dong-Sheng Xiong
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China
| | - Li Wang
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China
| | - Ya-Nan Zhang
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China
| | - Ling Yu
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China
| | - Jia-Jing Wei
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China
| | - Li-Bing He
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China
| | - Weiwei Zhi
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China
| | - Xin-Rong Du
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Ning-Jing Li
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Chang-Li Han
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - He-Qiu Yan
- School of Clinical Laboratory Medicine, Chengdu Medical College, Chengdu, 610083, China
| | - Zhuo-Ting Zhou
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Yang-Bao Miao
- Department of Haematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610000, China.
| | - Wen Wang
- Department of Haematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610000, China.
| | - Wei-Xin Liu
- School of Medicine and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China.
- Key Laboratory of Reproductive Medicine, Sichuan Provincial Maternity and Child Health Care Hospital, The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu, 610045, China.
| |
Collapse
|
6
|
Park S, Sharma H, Safdar M, Lee J, Kim W, Park S, Jeong HE, Kim J. Micro/nanoengineered agricultural by-products for biomedical and environmental applications. ENVIRONMENTAL RESEARCH 2024; 250:118490. [PMID: 38365052 DOI: 10.1016/j.envres.2024.118490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 02/08/2024] [Accepted: 02/13/2024] [Indexed: 02/18/2024]
Abstract
Agriculturally derived by-products generated during the growth cycles of living organisms as secondary products have attracted increasing interest due to their wide range of biomedical and environmental applications. These by-products are considered promising candidates because of their unique characteristics including chemical stability, profound biocompatibility and offering a green approach by producing the least impact on the environment. Recently, micro/nanoengineering based techniques play a significant role in upgrading their utility, by controlling their structural integrity and promoting their functions at a micro and nano scale. Specifically, they can be used for biomedical applications such as tissue regeneration, drug delivery, disease diagnosis, as well as environmental applications such as filtration, bioenergy production, and the detection of environmental pollutants. This review highlights the diverse role of micro/nano-engineering techniques when applied on agricultural by-products with intriguing properties and upscaling their wide range of applications across the biomedical and environmental fields. Finally, we outline the future prospects and remarkable potential that these agricultural by-products hold in establishing a new era in the realms of biomedical science and environmental research.
Collapse
Affiliation(s)
- Sunho Park
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea; Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea; Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, 61186, Republic of Korea; Department of Bio-Industrial Machinery Engineering, Pusan National University, Miryang, 50463, Republic of Korea
| | - Harshita Sharma
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea; Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea; Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Mahpara Safdar
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea; Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea; Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Jeongryun Lee
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea; Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea; Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Woochan Kim
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea; Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea; Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Sangbae Park
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea; Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea; Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, 61186, Republic of Korea; Department of Biosystems Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hoon Eui Jeong
- Department of Mechanical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea.
| | - Jangho Kim
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea; Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea; Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, 61186, Republic of Korea.
| |
Collapse
|
7
|
Li Z, Kang M, Xu C, Chiang M, Lee CS, Lee M. Black Phosphorus-Based Dynamic Self-Healing Hydrogel to Integrate Demineralized Bone Matrix and Noggin-Targeting siRNA for Synergistic Osteogenesis. ACS APPLIED MATERIALS & INTERFACES 2024:10.1021/acsami.4c01324. [PMID: 38686456 PMCID: PMC11522023 DOI: 10.1021/acsami.4c01324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
Although a demineralized bone matrix (DBM) is often used as an alternative to an autologous bone graft, its clinical application is still hampered by easy dispersion of DBM particles and insufficient osteoinductivity in the defect site. Herein, we designed a self-healing hydrogel for DBM that can rapidly restore its structural integrity after damage based on amino-rich black phosphorus (BP) nanosheets and aldehyde-functionalized hyaluronic acid (AHA). Given the increased expression of bone morphogenetic protein (BMP) antagonists by DBM stimulation, the osteogenic potency of DBM in the hydrogel carrier was further enhanced by abrogating the BMP antagonism. The BP/AHA hydrogel provided dynamic polymer-nanosheet networks that combine injectability, modability, and physical stability with high DBM loading, where the BP nanosheets served as osteogenic cross-linkers to promote biomineralization and deliver siRNA to suppress undesirable expression of BMP antagonist noggin by DBM. As a result, the BP/AHA hydrogel integrated with DBM and noggin-targeting siRNA synergistically promoted osteogenic differentiation of mesenchymal stem cells by enhancing BMP/Smad signaling. This work demonstrates a promising strategy to improve the efficacy of bone regeneration using bone graft.
Collapse
Affiliation(s)
- Zhi Li
- Division of Oral and Systemic Health Sciences, School of Dentistry, University of California, Los Angeles, CA 90095, United States
| | - Minjee Kang
- Division of Oral and Systemic Health Sciences, School of Dentistry, University of California, Los Angeles, CA 90095, United States
| | - Changlu Xu
- Division of Oral and Systemic Health Sciences, School of Dentistry, University of California, Los Angeles, CA 90095, United States
| | - Michelle Chiang
- Division of Oral and Systemic Health Sciences, School of Dentistry, University of California, Los Angeles, CA 90095, United States
| | - Chung-Sung Lee
- Department of Pharmaceutical Engineering, Soonchunhyang University, Asan 31538, Republic of Korea
| | - Min Lee
- Division of Oral and Systemic Health Sciences, School of Dentistry, University of California, Los Angeles, CA 90095, United States
- Department of Bioengineering, University of California, Los Angeles, CA 90095, United States
| |
Collapse
|
8
|
Reis IL, Lopes B, Sousa P, Sousa AC, Caseiro AR, Mendonça CM, Santos JM, Atayde LM, Alvites RD, Maurício AC. Equine Musculoskeletal Pathologies: Clinical Approaches and Therapeutical Perspectives-A Review. Vet Sci 2024; 11:190. [PMID: 38787162 PMCID: PMC11126110 DOI: 10.3390/vetsci11050190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 04/12/2024] [Accepted: 04/23/2024] [Indexed: 05/25/2024] Open
Abstract
Musculoskeletal injuries such as equine osteoarthritis, osteoarticular defects, tendonitis/desmitis, and muscular disorders are prevalent among sport horses, with a fair prognosis for returning to exercise or previous performance levels. The field of equine medicine has witnessed rapid and fruitful development, resulting in a diverse range of therapeutic options for musculoskeletal problems. Staying abreast of these advancements can be challenging, prompting the need for a comprehensive review of commonly used and recent treatments. The aim is to compile current therapeutic options for managing these injuries, spanning from simple to complex physiotherapy techniques, conservative treatments including steroidal and non-steroidal anti-inflammatory drugs, hyaluronic acid, polysulfated glycosaminoglycans, pentosan polysulfate, and polyacrylamides, to promising regenerative therapies such as hemoderivatives and stem cell-based therapies. Each therapeutic modality is scrutinized for its benefits, limitations, and potential synergistic actions to facilitate their most effective application for the intended healing/regeneration of the injured tissue/organ and subsequent patient recovery. While stem cell-based therapies have emerged as particularly promising for equine musculoskeletal injuries, a multidisciplinary approach is underscored throughout the discussion, emphasizing the importance of considering various therapeutic modalities in tandem.
Collapse
Affiliation(s)
- Inês L. Reis
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; (I.L.R.); (B.L.); (P.S.); (A.C.S.); (C.M.M.); (J.M.S.); (L.M.A.); (R.D.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
- Departamento de Ciências Veterinárias, Cooperativa de Ensino Superior Politécnico e Universitário (CESPU), Instituto Universitário de Ciências da Saúde (IUCS), Avenida Central de Gandra 1317, 4585-116 Gandra, Portugal
| | - Bruna Lopes
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; (I.L.R.); (B.L.); (P.S.); (A.C.S.); (C.M.M.); (J.M.S.); (L.M.A.); (R.D.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
| | - Patrícia Sousa
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; (I.L.R.); (B.L.); (P.S.); (A.C.S.); (C.M.M.); (J.M.S.); (L.M.A.); (R.D.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
| | - Ana C. Sousa
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; (I.L.R.); (B.L.); (P.S.); (A.C.S.); (C.M.M.); (J.M.S.); (L.M.A.); (R.D.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
| | - Ana R. Caseiro
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
- Veterinary Sciences Department, University School Vasco da Gama (EUVG), Avenida José R. Sousa Fernandes, Lordemão, 3020-210 Coimbra, Portugal
- Vasco da Gama Research Center (CIVG), University School Vasco da Gama (EUVG), Avenida José R. Sousa Fernandes, Lordemão, 3020-210 Coimbra, Portugal
| | - Carla M. Mendonça
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; (I.L.R.); (B.L.); (P.S.); (A.C.S.); (C.M.M.); (J.M.S.); (L.M.A.); (R.D.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
- Campus Agrário de Vairão, Centro Clínico de Equinos de Vairão (CCEV), Rua da Braziela n° 100, 4485-144 Vairão, Portugal
| | - Jorge M. Santos
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; (I.L.R.); (B.L.); (P.S.); (A.C.S.); (C.M.M.); (J.M.S.); (L.M.A.); (R.D.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
| | - Luís M. Atayde
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; (I.L.R.); (B.L.); (P.S.); (A.C.S.); (C.M.M.); (J.M.S.); (L.M.A.); (R.D.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
- Campus Agrário de Vairão, Centro Clínico de Equinos de Vairão (CCEV), Rua da Braziela n° 100, 4485-144 Vairão, Portugal
| | - Rui D. Alvites
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; (I.L.R.); (B.L.); (P.S.); (A.C.S.); (C.M.M.); (J.M.S.); (L.M.A.); (R.D.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
- Departamento de Ciências Veterinárias, Cooperativa de Ensino Superior Politécnico e Universitário (CESPU), Instituto Universitário de Ciências da Saúde (IUCS), Avenida Central de Gandra 1317, 4585-116 Gandra, Portugal
| | - Ana C. Maurício
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; (I.L.R.); (B.L.); (P.S.); (A.C.S.); (C.M.M.); (J.M.S.); (L.M.A.); (R.D.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
- Campus Agrário de Vairão, Centro Clínico de Equinos de Vairão (CCEV), Rua da Braziela n° 100, 4485-144 Vairão, Portugal
| |
Collapse
|
9
|
Angolkar M, Paramshetti S, Gahtani RM, Al Shahrani M, Hani U, Talath S, Osmani RAM, Spandana A, Gangadharappa HV, Gundawar R. Pioneering a paradigm shift in tissue engineering and regeneration with polysaccharides and proteins-based scaffolds: A comprehensive review. Int J Biol Macromol 2024; 265:130643. [PMID: 38467225 DOI: 10.1016/j.ijbiomac.2024.130643] [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: 10/13/2023] [Revised: 02/16/2024] [Accepted: 03/03/2024] [Indexed: 03/13/2024]
Abstract
In the realm of modern medicine, tissue engineering and regeneration stands as a beacon of hope, offering the promise of restoring form and function to damaged or diseased organs and tissues. Central to this revolutionary field are biological macromolecules-nature's own blueprints for regeneration. The growing interest in bio-derived macromolecules and their composites is driven by their environmentally friendly qualities, renewable nature, minimal carbon footprint, and widespread availability in our ecosystem. Capitalizing on these unique attributes, specific composites can be tailored and enhanced for potential utilization in the realm of tissue engineering (TE). This review predominantly concentrates on the present research trends involving TE scaffolds constructed from polysaccharides, proteins and glycosaminoglycans. It provides an overview of the prerequisites, production methods, and TE applications associated with a range of biological macromolecules. Furthermore, it tackles the challenges and opportunities arising from the adoption of these biomaterials in the field of TE. This review also presents a novel perspective on the development of functional biomaterials with broad applicability across various biomedical applications.
Collapse
Affiliation(s)
- Mohit Angolkar
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research (JSSAHER), Mysuru 570015, Karnataka, India
| | - Sharanya Paramshetti
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research (JSSAHER), Mysuru 570015, Karnataka, India
| | - Reem M Gahtani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha 61421, Saudi Arabia.
| | - Mesfer Al Shahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha 61421, Saudi Arabia.
| | - Umme Hani
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia.
| | - Sirajunisa Talath
- Department of Pharmaceutical Chemistry, RAK College of Pharmaceutical Sciences, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates.
| | - Riyaz Ali M Osmani
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research (JSSAHER), Mysuru 570015, Karnataka, India.
| | - Asha Spandana
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research (JSSAHER), Mysuru 570015, Karnataka, India.
| | | | - Ravi Gundawar
- Department of Pharmaceutical Quality Assurance, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India.
| |
Collapse
|
10
|
Vimalraj S, Govindarajan D, Sudhakar S, Suresh R, Palanivel P, Sekaran S. Chitosan derived chito-oligosaccharides promote osteoblast differentiation and offer anti-osteoporotic potential: Molecular and morphological evidence from a zebrafish model. Int J Biol Macromol 2024; 259:129250. [PMID: 38199551 DOI: 10.1016/j.ijbiomac.2024.129250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 12/15/2023] [Accepted: 01/03/2024] [Indexed: 01/12/2024]
Abstract
This study delves into the potential of chito-oligosaccharides (COS) to promote osteoblast differentiation and prevent osteoporosis, utilizing experiments with mouse MSCs and the zebrafish model. The preliminary biocompatibility study affirms the non-toxic nature of COS across various concentrations. In the osteoblast differentiation study, COS enhances ALP activity and calcium deposition at the cellular level. Moreover, COS induces the upregulation of molecular markers, including Runx2, Type I collagen, ALP, osteocalcin, and osteonectin in mouse MSCs. Zebrafish studies further demonstrate COS's anti-osteoporotic effects, showcasing its ability to expedite fin fracture repair, vertebral mineralization, and bone mineralization in dexamethasone-induced osteoporosis models. The scale regenerative study reveals that COS mitigates the detrimental effects of dexamethasone induced osteoclastic activity, reducing TRAP and hydroxyproline levels while elevating the expression of Runx2a MASNA isoform, collagen2α, OC, and ON mRNAs. Additionally, COS enhances calcium and phosphorus levels in regenerated scales, impacting the bone-healthy calcium-to‑phosphorus ratio. The study also suggests that COS modulates the MMP3-Osteopontin-MAPK signaling pathway. Overall, this comprehensive investigation underscores the potential of COS to prevent and treat osteoporosis. Its multifaceted cellular and molecular effects, combined with in vivo bone regeneration and repair, propose that COS may be effective in addressing osteoporosis and related bone disorders. Nonetheless, further research is imperative to unravel underlying mechanisms and optimize clinical applications.
Collapse
Affiliation(s)
- Selvaraj Vimalraj
- Department of Applied Mechanics and Biomedical Engineering, Indian Institute of Technology-Madras, Chennai 600 036, Tamil Nadu, India; Department of Prosthodontics, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Science (SIMATS), Saveetha University, Chennai 600 077, Tamil Nadu, India.
| | - Dharunya Govindarajan
- Department of Biotechnology, Stem Cell and Molecular Biology Laboratory, Bhupat & Jyoti Mehta School of Biosciences, Indian Institute of Technology, Madras, Chennai 600 036, Tamil Nadu, India
| | - Swathi Sudhakar
- Department of Applied Mechanics and Biomedical Engineering, Indian Institute of Technology-Madras, Chennai 600 036, Tamil Nadu, India
| | - Renugaa Suresh
- Department of Applied Mechanics and Biomedical Engineering, Indian Institute of Technology-Madras, Chennai 600 036, Tamil Nadu, India
| | | | - Saravanan Sekaran
- Department of Prosthodontics, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Science (SIMATS), Saveetha University, Chennai 600 077, Tamil Nadu, India
| |
Collapse
|
11
|
Mohanto S, Narayana S, Merai KP, Kumar JA, Bhunia A, Hani U, Al Fatease A, Gowda BHJ, Nag S, Ahmed MG, Paul K, Vora LK. Advancements in gelatin-based hydrogel systems for biomedical applications: A state-of-the-art review. Int J Biol Macromol 2023; 253:127143. [PMID: 37793512 DOI: 10.1016/j.ijbiomac.2023.127143] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/27/2023] [Accepted: 09/27/2023] [Indexed: 10/06/2023]
Abstract
A gelatin-based hydrogel system is a stimulus-responsive, biocompatible, and biodegradable polymeric system with solid-like rheology that entangles moisture in its porous network that gradually protrudes to assemble a hierarchical crosslinked arrangement. The hydrolysis of collagen directs gelatin construction, which retains arginyl glycyl aspartic acid and matrix metalloproteinase-sensitive degeneration sites, further confining access to chemicals entangled within the gel (e.g., cell encapsulation), modulating the release of encapsulated payloads and providing mechanical signals to the adjoining cells. The utilization of various types of functional tunable biopolymers as scaffold materials in hydrogels has become highly attractive due to their higher porosity and mechanical ability; thus, higher loading of proteins, peptides, therapeutic molecules, etc., can be further modulated. Furthermore, a stimulus-mediated gelatin-based hydrogel with an impaired concentration of gellan demonstrated great shear thinning and self-recovering characteristics in biomedical and tissue engineering applications. Therefore, this contemporary review presents a concise version of the gelatin-based hydrogel as a conceivable biomaterial for various biomedical applications. In addition, the article has recapped the multiple sources of gelatin and their structural characteristics concerning stimulating hydrogel development and delivery approaches of therapeutic molecules (e.g., proteins, peptides, genes, drugs, etc.), existing challenges, and overcoming designs, particularly from drug delivery perspectives.
Collapse
Affiliation(s)
- Sourav Mohanto
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, Karnataka, India.
| | - Soumya Narayana
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, Karnataka, India
| | - Khushboo Paresh Merai
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, Ahmedabad 382481, Gujrat, India
| | - Jahanvee Ashok Kumar
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, Ahmedabad 382481, Gujrat, India
| | - Adrija Bhunia
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, Karnataka, India
| | - Umme Hani
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia
| | - Adel Al Fatease
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia
| | - B H Jaswanth Gowda
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, Karnataka, India; School of Pharmacy, Queen's University Belfast, Medical Biology Centre, Belfast BT9 7BL, UK.
| | - Sagnik Nag
- Department of Bio-Sciences, School of Biosciences & Technology, Vellore Institute of Technology (VIT), Tiruvalam Rd, 632014, Tamil Nadu, India
| | - Mohammed Gulzar Ahmed
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, Karnataka, India
| | - Karthika Paul
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research (JSSAHER), Mysuru 570015, Karnataka, India
| | - Lalitkumar K Vora
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, Belfast BT9 7BL, UK
| |
Collapse
|
12
|
Xu Y, Zhuo J, Wang Q, Xu X, He M, Zhang L, Liu Y, Wu X, Luo K, Chen Y. Site-specific periosteal cells with distinct osteogenic and angiogenic characteristics. Clin Oral Investig 2023; 27:7437-7450. [PMID: 37848582 DOI: 10.1007/s00784-023-05333-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 10/10/2023] [Indexed: 10/19/2023]
Abstract
OBJECTIVES This study aimed to investigate the site-specific characteristics of rat mandible periosteal cells (MPCs) and tibia periosteal cells (TPCs) to assess the potential application of periosteal cells (PCs) in bone tissue engineering (BTE). MATERIALS AND METHODS MPCs and TPCs were isolated and characterized. The potential of proliferation, migration, osteogenesis and adipogenesis of MPCs and TPCs were evaluated by CCK-8, scratch assay, Transwell assay, alkaline phosphatase staining and activity, Alizarin Red S staining, RT‒qPCR, and Western blot (WB) assays, respectively. Then, these cells were cocultured with human umbilical vein endothelial cells (HUVECs) to investigate their angiogenic capacity, which was assessed by scratch assay, Transwell assay, Matrigel tube formation assay, RT‒qPCR, and WB assays. RESULTS MPCs exhibited higher osteogenic potential, higher alkaline phosphatase activity, and more mineralized nodule formation, while TPCs showed a greater capability for proliferation, migration, and adipogenesis. MPCs showed higher expression of angiogenic factors, and the conditioned medium of MPCs accelerated the migration of HUVECs, while MPC- conditioned medium induced the formation of more tubular structure in HUVECs in vitro. These data suggest that compared to TPCs, MPCs exert more consequential proangiogenic effects on HUVECs. CONCLUSIONS PCs possess skeletal site-specific differences in biological characteristics. MPCs exhibit more eminent osteogenic and angiogenic potentials, which highlights the potential application of MPCs for BTE. CLINICAL RELEVANCE Autologous bone grafting as the main modality for maxillofacial bone defect repair has many limitations. Constituting an important cell type in bone repair and regeneration, MPCs show greater potential for application in BTE, which provides a promising treatment option for maxillofacial bone defect repair.
Collapse
Affiliation(s)
- Yanmei Xu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, People's Republic of China
| | - Jin Zhuo
- Xuzhou Stomatological Hospital, Affiliated Stomatological Hospital of Xuzhou Medical University, Xuzhou, 221002, People's Republic of China
| | - Qisong Wang
- Longyan First Affiliated Hospital of Fujian Medical University, Longyan, 354000, People's Republic of China
| | - Xiongcheng Xu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, People's Republic of China
| | - Mengjiao He
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, People's Republic of China
| | - Lu Zhang
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, People's Republic of China
| | - Yijuan Liu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, People's Republic of China
| | - Xiaohong Wu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, People's Republic of China
| | - Kai Luo
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, People's Republic of China.
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, People's Republic of China.
| | - Yuling Chen
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, People's Republic of China.
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, People's Republic of China.
| |
Collapse
|
13
|
Wu MH, Wu K, Zhu YB, Li DC, Yang H, Zeng H. Baicalin Antagonizes Prostate Cancer Stemness via Inhibiting Notch1/NF-κB Signaling Pathway. Chin J Integr Med 2023; 29:914-923. [PMID: 37357241 DOI: 10.1007/s11655-023-3595-2] [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] [Accepted: 11/21/2022] [Indexed: 06/27/2023]
Abstract
OBJECTIVE To investigate the molecular mechanisms underlying the effect of baicalin on prostate cancer (PCa) progression both in vivo and in vitro. METHODS The in situ PCa stem cells (PCSCs)-injected xenograft tumor models were established in BALB/c nude mice. Tumor volume and weight were respectively checked after baicalin (100 mg/kg) treatment. Hematoxylin-eosin (HE) staining was used to observe the growth arrest and cell necrosis. mRNA expression levels of acetaldehyde dehydrogenase 1 (ALDH1), CD44, CD133 and Notch1 were determined by reverse transcription-polymerase chain reaction. Protein expression levels of ALDH1, CD44, CD133, Notch1, nuclear factor κB (NF-κB) P65 and NF-κB p-P65 were detected by Western blot. Expression and subcellular location of ALDH1, CD44, CD133, Notch1 and NF-κB p65 were detected by immunofluorescence analysis. In vitro, cell cycle distribution and cell apoptosis of PC3 PCSCs was assessed by flow cytometry after baicalin (125 µmol/L) treatment. The migration and invasion abilities of PCSCs were assessed using Transwell assays. Transmission electron microscopy scanning was utilized to observe the structure and autophagosome formation of baicalin-treated PCSCs. In addition, PCSCs were infected with lentiviruses expressing human Notch1. RESULTS Compared with the control group, the tumor volume and weight were notably reduced in mice treated with 100 mg/kg baicalin (P<0.05 or P<0.01). Histopathological analysis showed that baicalin treatment significantly inhibited cell proliferation and promoted cell apoptosis. Furthermore, baicalin treatment reduced mRNA and protein expression levels of CD44, CD133, ALDH1, and Notch1 as well as the protein expression of NF-κB p-P65 in the xenograft tumor (P<0.01). In vitro, the cell proliferation of PCSCs was significantly attenuated after treatment with 125 µmol/L baicalin for 72 h (P<0.01). The cell migration and invasion rates were decreased following treatment with baicalin for 48 and 72 h (P<0.01). Baicalin notably induced cell apoptosis and seriously damaged the structure of PCSCs. The mRNA and protein expressions of CD133, CD44, ALDH1 and Notch1 in PCSCs were significantly downregulated following baicalin treatment (P<0.01). Importantly, the inhibitory effects of baicalin on PCa progression and stemness were reversed by Notch1 overexpression (P<0.05 or P<0.01). CONCLUSION Mechanistically, baicalin exhibited a potential therapeutic effect on PCa via inhibiting the Notch1/NF-κB signaling pathway and its mediated cancer stemness.
Collapse
Affiliation(s)
- Ming-Hui Wu
- Department of Urology, Hubei University of Traditional Chinese Medicine, Wuhan, 430065, China
| | - Kun Wu
- Department of Urology, Hubei University of Traditional Chinese Medicine, Wuhan, 430065, China
| | - Yuan-Bing Zhu
- Department of Urology, Chongqing Jiangjin District Hospital of Chinese Medicine, Chongqing, 402260, China
| | - Da-Chuan Li
- Department of Urology, Chongqing Jiangjin District Hospital of Chinese Medicine, Chongqing, 402260, China
| | - Huan Yang
- Department of Urology, Chongqing Jiangjin District Hospital of Chinese Medicine, Chongqing, 402260, China
| | - Hong Zeng
- Department of Urology, Chongqing Jiangjin District Hospital of Chinese Medicine, Chongqing, 402260, China.
| |
Collapse
|
14
|
Zhu G, Zhou Y, Xu Y, Wang L, Han M, Xi K, Tang J, Li Z, Kou Y, Zhou X, Feng Y, Gu Y, Chen L. Functionalized acellular periosteum guides stem cell homing to promote bone defect repair. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2023; 34:2000-2020. [PMID: 37071056 DOI: 10.1080/09205063.2023.2204779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 04/17/2023] [Indexed: 04/19/2023]
Abstract
The periosteum plays a key role in bone tissue regeneration, especially in the promotion and protection of new bones. However, among the bone repair materials, many biomimetic artificial periosteum lack the natural periosteal structure, stem cells, and immunoregulation required for bone regeneration. In this study, we used natural periosteum to produce acellular periosteum. To retain the appropriate cell survival structure and immunomodulatory proteins, we grafted the functional polypeptide SKP on the surface collagen of the periosteum via an amide bond, providing the acellular periosteum with the ability to recruit mesenchymal stem cells. Thus, we developed a biomimetic periosteum (DP-SKP) with the ability to promote stem cell homing and immunoregulation in vivo. Compared to the blank and simple decellularized periosteum groups, DP-SKP was more conducive to stem cell adhesion, growth, and osteogenic differentiation in vitro. Additionally, compared with the other two groups, DP-SKP significantly promoted mesenchymal stem cell homing to the periosteal transplantation site, improved the bone immune microenvironment, and accelerated new lamellar bone formation in the critical size defect of rabbit skulls in vivo. Therefore, this acellular periosteum with a mesenchymal stem cell homing effect is expected to be used as an extracellular artificial periosteum in clinical practice.
Collapse
Affiliation(s)
- Guoqing Zhu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Orthopedics, Suzhou Municipal Hospital, Suzhou, China
| | - Yidi Zhou
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yichang Xu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Lingjun Wang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Meng Han
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Orthopedics, Xuzhou Central Hospital, Xuzhou, China
| | - Kun Xi
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jincheng Tang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ziang Li
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yu Kou
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xindie Zhou
- Department of Orthopedics, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China
| | - Yu Feng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yong Gu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Liang Chen
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
15
|
Rajabi M, Cabral JD, Saunderson S, Ali MA. 3D printing of chitooligosaccharide-polyethylene glycol diacrylate hydrogel inks for bone tissue regeneration. J Biomed Mater Res A 2023; 111:1468-1481. [PMID: 37066870 DOI: 10.1002/jbm.a.37548] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 03/09/2023] [Accepted: 04/03/2023] [Indexed: 04/18/2023]
Abstract
To date, lack of functional hydrogel inks has limited 3D printing applications in tissue engineering. This study developed a series of photocurable hydrogel inks based on chitooligosaccharide (COS)-polyethylene glycol diacrylate (PEGDA) for extrusion-based 3D printing of bone tissue scaffolds. The scaffolds were prepared by aza-Michael addition of COS and PEGDA followed by photopolymerisation of unreacted PEGDA. The hydrogel inks showed sufficient shear thinning properties required for extrusion 3D printing. The printed scaffolds exhibited excellent shape fidelity and fine microstructure with a resolution of 250 μm. By increasing the COS content, the swelling ratio of the scaffolds decreased, while the compressive strength increased. 3D printed COS-PEGDA scaffolds showed high viability of human bone mesenchymal stem cells in vitro. In addition, scaffolds containing 2 wt% COS showed significantly higher alkaline phosphatase activity, calcium deposition, and bioactivity in simulated body fluid compared to the control (PEGDA). Altogether, 3D printed COS-PEGDA scaffolds represent promising candidates for bone tissue regeneration.
Collapse
Affiliation(s)
- Mina Rajabi
- Faculty of Dentistry, Division of Health Sciences, Centre for Bioengineering & Nanomedicine, Sir John Walsh Research Institute, University of Otago, Dunedin, New Zealand
| | - Jaydee D Cabral
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Sarah Saunderson
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - M Azam Ali
- Faculty of Dentistry, Division of Health Sciences, Centre for Bioengineering & Nanomedicine, Sir John Walsh Research Institute, University of Otago, Dunedin, New Zealand
| |
Collapse
|
16
|
Roldan L, Isaza C, Ospina J, Montoya C, Domínguez J, Orrego S, Correa S. A Comparative Study of HA/DBM Compounds Derived from Bovine and Porcine for Bone Regeneration. J Funct Biomater 2023; 14:439. [PMID: 37754853 PMCID: PMC10532284 DOI: 10.3390/jfb14090439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 07/25/2023] [Accepted: 08/17/2023] [Indexed: 09/28/2023] Open
Abstract
This comparative study investigated the tissue regeneration and inflammatory response induced by xenografts comprised of hydroxyapatite (HA) and demineralized bone matrix (DBM) extracted from porcine (P) and bovine (B) sources. First, extraction of HA and DBM was independently conducted, followed by chemical and morphological characterization. Second, mixtures of HA/DBM were prepared in 50/50 and 60/40 concentrations, and the chemical, morphological, and mechanical properties were evaluated. A rat calvarial defect model was used to evaluate the tissue regeneration and inflammatory responses at 3 and 6 months. The commercial allograft DBM Puros® was used as a clinical reference. Different variables related to tissue regeneration were evaluated, including tissue thickness regeneration (%), amount of regenerated bone area (%), and amount of regenerated collagen area (%). The inflammatory response was evaluated by quantifying the blood vessel area. Overall, tissue regeneration from porcine grafts was superior to bovine. After 3 months of implantation, the tissue thickness regeneration in the 50/50P compound and the commercial DBM was significantly higher (~99%) than in the bovine materials (~23%). The 50/50P and DBM produced higher tissue regeneration than the naturally healed controls. Similar trends were observed for the regenerated bone and collagen areas. The blood vessel area was correlated with tissue regeneration in the first 3 months of evaluation. After 6 months of implantation, HA/DBM compounds showed less regenerated collagen than the DBM-only xenografts. In addition, all animal-derived xenografts improved tissue regeneration compared with the naturally healed defects. No clinical complications associated with any implanted compound were noted.
Collapse
Affiliation(s)
- Lina Roldan
- Grupo de Investigación en Bioingeniería (GIB), Universidad EAFIT, Medellín 050022, Colombia; (L.R.); (C.I.)
- Department of Oral Health Sciences, Kornberg School of Dentistry, Temple University, Philadelphia, PA 19122, USA; (C.M.); (S.O.)
| | - Catalina Isaza
- Grupo de Investigación en Bioingeniería (GIB), Universidad EAFIT, Medellín 050022, Colombia; (L.R.); (C.I.)
| | - Juan Ospina
- Centro de Investigación y Desarrollo Cárnico, Industrias de Alimentos Zenú S.A.S., Grupo Nutresa, Medellín 050044, Colombia;
| | - Carolina Montoya
- Department of Oral Health Sciences, Kornberg School of Dentistry, Temple University, Philadelphia, PA 19122, USA; (C.M.); (S.O.)
| | - José Domínguez
- Grupo de Investigación en Bioingeniería (GIB), Universidad EAFIT, Medellín 050022, Colombia; (L.R.); (C.I.)
| | - Santiago Orrego
- Department of Oral Health Sciences, Kornberg School of Dentistry, Temple University, Philadelphia, PA 19122, USA; (C.M.); (S.O.)
- Bioengineering Department, College of Engineering, Temple University, Philadelphia, PA 191122, USA
| | - Santiago Correa
- Grupo de Investigación en Bioingeniería (GIB), Universidad EAFIT, Medellín 050022, Colombia; (L.R.); (C.I.)
- Escuela de Ciencias Aplicadas e Ingeniería, Universidad EAFIT, Medellín 050022, Colombia
| |
Collapse
|
17
|
Vedakumari SW, Jancy SJV, Prabakaran L, Raja Pravin Y, Senthil R. A review on background, process and application of electrospun nanofibers for tissue regeneration. Proc Inst Mech Eng H 2023:9544119231164713. [PMID: 37060196 DOI: 10.1177/09544119231164713] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2023]
Abstract
Electrospinning is a versatile method which is used to synthesize nano/micro sized fibers under the influence of electric field. Electrospun nanoscaffolds are one of the widely accepted platforms for cultivating soft and hard tissues as they create a prefect micro-environment for cell adhesion, proliferation and differentiation. Nanoscaffolds are widely used in the field of tissue engineering due to their versatility in aiding the growth of different types of cells and tissues for varied applications. The composition, molecular weight and structure of polymer used to fabricate nanoscaffold plays an important role in determining the size and strength of the nanofibers prepared. This review gives information about the background, process and different types of polymers used in electrospinning. Recent advances in culturing liver cells, osteoblasts, skin cells, neural cells and coronary artery smooth muscle cells on nanoscaffolds are also elucidated.
Collapse
Affiliation(s)
- Sathyaraj Weslen Vedakumari
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chennai, Tamil Nadu, India
| | - Sathyaraj Jacqulin Veda Jancy
- Department of Computer and Communication Engineering, Sri Sai Ram Institute of Technology, Chennai, Tamil Nadu, India
| | - Lokesh Prabakaran
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chennai, Tamil Nadu, India
- National Centre for Nanoscience and Nanotechnology, University of Madras, Guindy Campus, Chennai, Tamil Nadu, India
| | - Yovan Raja Pravin
- Department of Physics (Science and Humanities), Agni College of Technology, Chennai, Tamil Nadu, India
| | - Rethinam Senthil
- Department of Leather Engineering, Faculty of Engineering, Ege University, Bornova, Izmir, Turkey
- School of Bio & Chemical Engineering, Sathyabama University, Chennai, Tamil Nadu, India
- Department of Pharmacology, Saveetha Dental College and Hospitals, SIMATS, Chennai, Tamil Nadu, India
| |
Collapse
|
18
|
李 晶, 任 晓, 乔 玮, 石 浩, 杨 婷, 马 绍, 李 宝, 赵 亚. [Preparation and bone repair capability of a new plastic bone filler material]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2023; 37:302-307. [PMID: 36940988 PMCID: PMC10027535 DOI: 10.7507/1002-1892.202210022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 02/08/2023] [Indexed: 03/23/2023]
Abstract
Objective To prepare a new plastic bone filler material with adhesive carrier and matrix particles derived from human bone, and evaluate its safety and osteoinductive ability through animal tests. Methods The human long bones donated voluntarily were prepared into decalcified bone matrix (DBM) by crushing, cleaning, and demineralization, and then the DBM was prepared into bone matrix gelatin (BMG) by warm bath method, and the BMG and DBM were mixed to prepare the experimental group's plastic bone filler material; DBM was used as control group. Fifteen healthy male thymus-free nude mice aged 6-9 weeks were used to prepare intermuscular space between gluteus medius and gluteus maximus muscles, and all of them were implanted with experimental group materials. The animals were sacrificed at 1, 4, and 6 weeks after operation, and the ectopic osteogenic effect was evaluated by HE staining. Eight 9-month-old Japanese large-ear rabbits were selected to prepare 6-mm-diameter defects at the condyles of both hind legs, and the left and right sides were filled with the materials of the experimental group and the control group respectively. The animals were sacrificed at 12 and 26 weeks after operation, and the effect of bone defect repair were evaluated by Micro-CT and HE staining. Results In ectopic osteogenesis experiment, HE staining showed that a large number of chondrocytes could be observed at 1 week after operation, and obvious newly formed cartilage tissue could be observed at 4 and 6 weeks after operation. For the rabbit condyle bone filling experiment, HE staining showed that at 12 weeks after operation, part of the materials were absorbed, and new cartilage could be observed in both experimental and control groups; at 26 weeks after operation, the most of the materials were absorbed, and large amount of new bone could be observed in the 2 groups, while new bone unit structure could be observed in the experimental group. Micro-CT observation showed that the bone formation rate and area of the experimental group were better than those of the control group. The measurement of bone morphometric parameters showed that the parameters at 26 weeks after operation in both groups were significantly higher than those at 12 weeks after operation ( P<0.05). At 12 weeks after operation, the bone mineral density and bone volume fraction in the experimental group were significantly higher than those in the control group ( P<0.05), and there was no significant difference between the two groups in trabecular thickness ( P>0.05). At 26 weeks after operation, the bone mineral density of the experimental group was significantly higher than that of the control group ( P<0.05). There was no significant difference in bone volume fraction and trabecular thickness between the two groups ( P>0.05). Conclusion The new plastic bone filler material is an excellent bone filler material with good biosafety and osteoinductive activity.
Collapse
Affiliation(s)
- 晶 李
- 中国辐射防护研究院生物材料研发中心 山西奥瑞生物材料有限公司(太原 030006)Biomaterial R&D Center of China Institute for Radiation Protection, Shanxi Osteorad Biomaterial Co., Ltd, Taiyuan Shanxi, 030006, P. R. China
| | - 晓琦 任
- 中国辐射防护研究院生物材料研发中心 山西奥瑞生物材料有限公司(太原 030006)Biomaterial R&D Center of China Institute for Radiation Protection, Shanxi Osteorad Biomaterial Co., Ltd, Taiyuan Shanxi, 030006, P. R. China
| | - 玮 乔
- 中国辐射防护研究院生物材料研发中心 山西奥瑞生物材料有限公司(太原 030006)Biomaterial R&D Center of China Institute for Radiation Protection, Shanxi Osteorad Biomaterial Co., Ltd, Taiyuan Shanxi, 030006, P. R. China
| | - 浩 石
- 中国辐射防护研究院生物材料研发中心 山西奥瑞生物材料有限公司(太原 030006)Biomaterial R&D Center of China Institute for Radiation Protection, Shanxi Osteorad Biomaterial Co., Ltd, Taiyuan Shanxi, 030006, P. R. China
| | - 婷 杨
- 中国辐射防护研究院生物材料研发中心 山西奥瑞生物材料有限公司(太原 030006)Biomaterial R&D Center of China Institute for Radiation Protection, Shanxi Osteorad Biomaterial Co., Ltd, Taiyuan Shanxi, 030006, P. R. China
| | - 绍英 马
- 中国辐射防护研究院生物材料研发中心 山西奥瑞生物材料有限公司(太原 030006)Biomaterial R&D Center of China Institute for Radiation Protection, Shanxi Osteorad Biomaterial Co., Ltd, Taiyuan Shanxi, 030006, P. R. China
| | - 宝兴 李
- 中国辐射防护研究院生物材料研发中心 山西奥瑞生物材料有限公司(太原 030006)Biomaterial R&D Center of China Institute for Radiation Protection, Shanxi Osteorad Biomaterial Co., Ltd, Taiyuan Shanxi, 030006, P. R. China
| | - 亚平 赵
- 中国辐射防护研究院生物材料研发中心 山西奥瑞生物材料有限公司(太原 030006)Biomaterial R&D Center of China Institute for Radiation Protection, Shanxi Osteorad Biomaterial Co., Ltd, Taiyuan Shanxi, 030006, P. R. China
| |
Collapse
|
19
|
The differentiation of human induced pluripotent stem cells into hematopoietic stem cells on 3D bone scaffold in a dynamic culture system. Tissue Cell 2023; 82:102044. [PMID: 36905860 DOI: 10.1016/j.tice.2023.102044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 02/16/2023] [Accepted: 02/16/2023] [Indexed: 02/19/2023]
Abstract
Hematopoietic stem cell transplantation is used for cell-based therapy for many hematological disorders. However, difficulty in finding proper donors has limited this source of stem cells. For clinical application, the generation of these cells from induced pluripotent stem cells (iPSs) is a fascinating and endless source. One of the experimental methods to generate HSCs from iPSs is the mimicking of the hematopoietic niche. In the current study, as the first phase of differentiation, embryoid bodies were formed from iPSs. They were then cultured in different dynamic conditions in order to determine the appropriate settings for their differentiation into HSCs. The dynamic culture was composed of DBM Scaffold with or without growth factor. After ten days, the specific HSC markers (CD34, CD133, CD31 and CD45) were assessed using flow-cytometry. Our findings demonstrated that the dynamic conditions were significantly suitable than static ones. In addition, in 3D scaffold and dynamic system the expression of CXCR4, as a homing marker, was increased. These results suggest that the 3D culture bioreactor with DBM scaffold could provide a new approach for differentiation of iPSs into HSCs. Moreover, this system could provide maximum mimicry of bone marrow niche.
Collapse
|
20
|
Hatt LP, Armiento AR, Mys K, Thompson K, Hildebrand M, Nehrbass D, Müller WEG, Zeiter S, Eglin D, Stoddart MJ. Standard in vitro evaluations of engineered bone substitutes are not sufficient to predict in vivo preclinical model outcomes. Acta Biomater 2023; 156:177-189. [PMID: 35988660 DOI: 10.1016/j.actbio.2022.08.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 08/10/2022] [Accepted: 08/11/2022] [Indexed: 01/18/2023]
Abstract
Understanding the optimal conditions required for bone healing can have a substantial impact to target the problem of non-unions and large bone defects. The combination of bioactive factors, regenerative progenitor cells and biomaterials to form a tissue engineered (TE) complex is a promising solution but translation to the clinic has been slow. We hypothesized the typical material testing algorithm used is insufficient and leads to materials being mischaracterized as promising. In the first part of this study, human bone marrow - derived mesenchymal stromal cells (hBM-MSCs) were embedded in three commonly used biomaterials (hyaluronic acid methacrylate, gelatin methacrylate and fibrin) and combined with relevant bioactive osteogenesis factors (dexamethasone microparticles and polyphosphate nanoparticles) to form a TE construct that underwent in vitro osteogenic differentiation for 28 days. Gene expression of relevant transcription factors and osteogenic markers, and von Kossa staining were performed. In the second and third part of this study, the same combination of TE constructs were implanted subcutaneously (cell containing) in T cell-deficient athymic Crl:NIH-Foxn1rnu rats for 8 weeks or cell free in an immunocompetent New Zealand white rabbit calvarial model for 6 weeks, respectively. Osteogenic performance was investigated via MicroCT imaging and histology staining. The in vitro study showed enhanced upregulation of relevant genes and significant mineral deposition within the three biomaterials, generally considered as a positive result. Subcutaneous implantation indicates none to minor ectopic bone formation. No enhanced calvarial bone healing was detected in implanted biomaterials compared to the empty defect. The reasons for the poor correlation of in vitro and in vivo outcomes are unclear and needs further investigation. This study highlights the discrepancy between in vitro and in vivo outcomes, demonstrating that in vitro data should be interpreted with extreme caution. In vitro models with higher complexity are necessary to increase value for translational studies. STATEMENT OF SIGNIFICANCE: Preclinical testing of newly developed biomaterials is a crucial element of the development cycle. Despite this, there is still significant discrepancy between in vitro and in vivo test results. Within this study we investigate multiple combinations of materials and osteogenic stimulants and demonstrate a poor correlation between the in vitro and in vivo data. We propose rationale for why this may be the case and suggest a modified testing algorithm.
Collapse
Affiliation(s)
- Luan P Hatt
- AO Research Institute Davos, 7270 Davos Platz, Switzerland; Institute for Biomechanics, ETH Zürich; 8093 Zürich, Switzerland
| | | | - Karen Mys
- AO Research Institute Davos, 7270 Davos Platz, Switzerland
| | - Keith Thompson
- AO Research Institute Davos, 7270 Davos Platz, Switzerland
| | | | - Dirk Nehrbass
- AO Research Institute Davos, 7270 Davos Platz, Switzerland
| | - Werner E G Müller
- Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Stephan Zeiter
- AO Research Institute Davos, 7270 Davos Platz, Switzerland
| | - David Eglin
- Mines Saint-Etienne, Univ Lyon, Univ Jean Monnet, INSERM, U 1059 Sainbiose, Centre CIS, F-42023 Saint-Etienne, France
| | | |
Collapse
|
21
|
Sun D, Mou S, Chen L, Yang J, Wang R, Zhong A, Wang W, Tong J, Wang Z, Sun J. High yield engineered nanovesicles from ADSC with enriched miR-21-5p promote angiogenesis in adipose tissue regeneration. Biomater Res 2022; 26:83. [PMID: 36528594 PMCID: PMC9758932 DOI: 10.1186/s40824-022-00325-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 11/18/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) have been found to have a great potential for soft tissue repair due to various biological functions, including pro-angiogenesis and low immunogenicity. However, the low yield and heterogeneity of MSC-EVs limited their clinical transformation. This study was designed to develop a novel adipose-derived stem cell engineered nanovesicles (ADSC-NVs) with high production and explore its pro-angiogenetic effect and application in adipose tissue regeneration. METHODS Adipose-derived stem cell-derived extracellular vesicles (ADSC-EVs) were isolated from an EVs-free culture medium for human ADSCs (hADSCs). ADSC-NVs were prepared by sequentially extruding ADSCs followed by iodixanol density gradient ultracentrifugation and were compared with ADSC-EVs in morphology, size distribution, protein contents and yield. The pro-angiogenetic effect of ADSC-NVs in different doses (0, 5, 20 and 80 μg/mL) in vitro was determined using transwell assay, tube formation assay, western blot and qRT-PCR. In vivo, BALB/c nude mice were administered injection of a mixture of fat granules and different dose of ADSC-NVs and grafts were harvested at 12 weeks post-transplantation for further analysis. By analyzing the weight and volume of grafts and histological evaluation, we investigated the effect of ADSC-NVs in vessel formation and adipose tissue regeneration. RESULTS Our results showed yield of purified ADSC-NVs was approximately 20 times more than that of ADSC-EVs secreted by the same number of ADSCs. In vitro, both ADSC-NVs and ADSC-EVs exhibited a dose-dependent pro-angiogenetic effect, despite their distinct miRNA profiles. These effects of ADSC-NVs may be mediated by enriched miR-21-5p via PTEN inhibition and PI3K/p-Akt signaling activation. Furthermore, after a mixed injection of ADSC-NVs, vessel formation and adipose regeneration were observed in vivo in fat implants. CONCLUSIONS Our study developed a potent alternative of ADSC-EVs. ADSC-NVs have a high pro-angiogenesis potential and can be used as cell-free therapeutic biomaterials in soft tissue regeneration.
Collapse
Affiliation(s)
- Di Sun
- grid.33199.310000 0004 0368 7223Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China ,Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022 China
| | - Shan Mou
- grid.33199.310000 0004 0368 7223Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China ,Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022 China
| | - Lifeng Chen
- grid.33199.310000 0004 0368 7223Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China ,Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022 China
| | - Jie Yang
- grid.33199.310000 0004 0368 7223Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China ,Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022 China
| | - Rongrong Wang
- grid.33199.310000 0004 0368 7223Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China ,Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022 China
| | - Aimei Zhong
- grid.33199.310000 0004 0368 7223Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China ,Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022 China
| | - Wei Wang
- grid.33199.310000 0004 0368 7223Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China ,Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022 China
| | - Jing Tong
- grid.33199.310000 0004 0368 7223Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China ,Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022 China
| | - Zhenxing Wang
- grid.33199.310000 0004 0368 7223Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China ,Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022 China
| | - Jiaming Sun
- grid.33199.310000 0004 0368 7223Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China ,Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022 China
| |
Collapse
|
22
|
Bhushan S, Singh S, Maiti TK, Sharma C, Dutt D, Sharma S, Li C, Tag Eldin EM. Scaffold Fabrication Techniques of Biomaterials for Bone Tissue Engineering: A Critical Review. Bioengineering (Basel) 2022; 9:728. [PMID: 36550933 PMCID: PMC9774188 DOI: 10.3390/bioengineering9120728] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/17/2022] [Accepted: 09/20/2022] [Indexed: 11/27/2022] Open
Abstract
Bone tissue engineering (BTE) is a promising alternative to repair bone defects using biomaterial scaffolds, cells, and growth factors to attain satisfactory outcomes. This review targets the fabrication of bone scaffolds, such as the conventional and electrohydrodynamic techniques, for the treatment of bone defects as an alternative to autograft, allograft, and xenograft sources. Additionally, the modern approaches to fabricating bone constructs by additive manufacturing, injection molding, microsphere-based sintering, and 4D printing techniques, providing a favorable environment for bone regeneration, function, and viability, are thoroughly discussed. The polymers used, fabrication methods, advantages, and limitations in bone tissue engineering application are also emphasized. This review also provides a future outlook regarding the potential of BTE as well as its possibilities in clinical trials.
Collapse
Affiliation(s)
- Sakchi Bhushan
- Department of Paper Technology, IIT Roorkee, Saharanpur 247001, India
| | - Sandhya Singh
- Department of Paper Technology, IIT Roorkee, Saharanpur 247001, India
| | - Tushar Kanti Maiti
- Department of Polymer and Process Engineering, IIT Roorkee, Saharanpur 247001, India
| | - Chhavi Sharma
- Department of Polymer and Process Engineering, IIT Roorkee, Saharanpur 247001, India
| | - Dharm Dutt
- Department of Paper Technology, IIT Roorkee, Saharanpur 247001, India
| | - Shubham Sharma
- Mechanical Engineering Department, University Center for Research & Development, Chandigarh University, Mohali 140413, India
- School of Mechanical and Automotive Engineering, Qingdao University of Technology, Qingdao 266520, China
| | - Changhe Li
- School of Mechanical and Automotive Engineering, Qingdao University of Technology, Qingdao 266520, China
| | | |
Collapse
|
23
|
Stukel Shah JM, Lundquist B, Macaitis J, Pfau-Cloud MR, Beltran FO, Grunlan MA, Lien W, Wang HC, Burdette AJ. Comparative evaluation of mesenchymal stromal cell growth and osteogenic differentiation on a shape memory polymer scaffold. J Biomed Mater Res B Appl Biomater 2022; 110:2063-2074. [PMID: 35344262 DOI: 10.1002/jbm.b.35061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 03/08/2022] [Accepted: 03/12/2022] [Indexed: 11/06/2022]
Abstract
Trauma-induced, critical-size bone defects pose a clinical challenge to heal. Albeit autografts are the standard-of-care, they are limited by their inability to be shaped to various defect geometries and often incur donor site complications. Herein, the combination of a "self-fitting" shape memory polymer (SMP) scaffold and seeded mesenchymal stromal cells (MSCs) was investigated as an alternative. The porous SMP scaffold, prepared from poly(ε-caprolactone) diacrylate (PCL-DA) and coated with polydopamine, provided conformal shaping and cell adhesion. MSCs from five tissues, amniotic (AMSCs), chorionic tissue (CHSCs), umbilical cord (UCSCs), adipose (ADSCs), and bone marrow (BMSCs) were evaluated for viability, density, and osteogenic differentiation on the SMP scaffold. BMSCs exhibited the fastest increase in cell density by day 3, but after day 10, CHSCs, UCSCs, and ADSCs approached similar cell density. BMSCs also showed the greatest calcification among the cell types, followed closely by ADSCs, CHSCs and AMSCs. Alkaline phosphatase (ALP) activity peaked at day 7 for AMSCs, UCSCs, ADSCs and BMSCs, and at day 14 for CHSCs, which had the greatest overall ALP activity. Of all the cell types, only scaffolds cultured with ADSCs in osteogenic media had increased hardness and local modulus as compared to blank scaffolds after 21 days of cell culture and osteogenic differentiation. Overall, ADSCs performed most favorably on the SMP scaffold. The SMP scaffold was able to support key cellular behaviors of MSCs and could potentially be a viable, regenerative alternative to autograft.
Collapse
Affiliation(s)
- Jessica M Stukel Shah
- Directorate of Combat Casualty Care & Operational Medicine, Naval Medical Research Unit San Antonio, San Antonio, Texas, USA
| | - Bridney Lundquist
- Directorate of Combat Casualty Care & Operational Medicine, Naval Medical Research Unit San Antonio, San Antonio, Texas, USA
| | - Joseph Macaitis
- Directorate of Combat Casualty Care & Operational Medicine, Naval Medical Research Unit San Antonio, San Antonio, Texas, USA
| | - Michaela R Pfau-Cloud
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - Felipe O Beltran
- Department of Materials Science and Engineering, Texas A&M University, College Station, Texas, USA
| | - Melissa A Grunlan
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA.,Department of Materials Science and Engineering, Texas A&M University, College Station, Texas, USA.,Department of Chemistry, Texas A&M University, College Station, Texas, USA
| | - Wen Lien
- USAF Dental Research & Consultation Service, JBSA Fort Sam Houston, San Antonio, Texas, USA
| | - Heuy-Ching Wang
- Directorate of Combat Casualty Care & Operational Medicine, Naval Medical Research Unit San Antonio, San Antonio, Texas, USA
| | - Alexander J Burdette
- Directorate of Combat Casualty Care & Operational Medicine, Naval Medical Research Unit San Antonio, San Antonio, Texas, USA
| |
Collapse
|
24
|
Dash S, . P, Arora V, Sachdeva K, Sharma H, Dinda AK, Agrawal AK, Jassal M, Mohanty S. Promoting in-vivo bone regeneration using facile engineered load-bearing 3D bioactive scaffold. Biomed Mater 2022. [DOI: 10.1088/1748-605x/ac58d6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Abstract
The worldwide incidence of bone disorders has trended steeply upward and is expected to get doubled by 2030. Biological mechanism of bone repair involves both osteo-conductivity and osteo-inductivity. In spite of the self-healing functionality after injury, bone tissue faces multitude of pathological challenges. Several innovative approaches have been developed to prepare biomaterial-based bone grafts. To design a suitable bone material, the freeze-drying technique has achieved significant importance among the other conventional methods. However, the functionality of the polymeric freeze-dried scaffold in in-vivo osteogenesis is in nascent stage. In this study facile, freeze dried, biomaterial-based load bearing 3D porous composite scaffolds have been prepared. The biocompatible scaffolds have been made by using chitosan (C), polycaprolactone (P), hydroxyapatite (H), glass ionomer (G), and graphene (gr). Scaffolds of eight different groups (C, P, CP, CPH, CPHG, CPHGgr1, CPHGgr2, CPHGgr3) have been designed and characterized to evaluate their applicability in orthopaedics. To evaluate the efficacy of the scaffolds a series of physio-chemical, morphological, and in-vitro & in-vivo biological experiments have been performed. From the obtained results it was observed that the CPHGgr1 is the ideal compatible material for Wharton’s Jelly derived mesenchymal stem cells and the blood cells. The in-vitro bone specific gene expression study revealed that, the scaffold assists MSCs osteogenic differentiation. Additionally, the in-vivo study on mice model was also performed for a period of 4 weeks and 8 weeks. The sub-cutaneous implantation of the designed scaffolds didn’t show any altered physiological condition in the animals, which indicated the in-vivo biocompatibility of the designed material. The histopathological study revealed that after 8 weeks of implantation, the CPHGgr1 scaffold supported significantly better collagen deposition and calcification. The facile designing of CPHGgr1 multicomponent nanocomposite provided an osteo-regenerative biomaterial with desired mechanical strength as an ideal regenerative material for cancellous bone tissue regeneration.
Collapse
|
25
|
Mineralization in a Critical Size Bone-Gap in Sheep Tibia Improved by a Chitosan-Calcium Phosphate-Based Composite as Compared to Predicate Device. MATERIALS 2022; 15:ma15030838. [PMID: 35160784 PMCID: PMC8836995 DOI: 10.3390/ma15030838] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/18/2022] [Accepted: 01/20/2022] [Indexed: 02/04/2023]
Abstract
Deacetylated chitin derivatives have been widely studied for tissue engineering purposes. This study aimed to compare the efficacy of an injectable product containing a 50% deacetylated chitin derivative (BoneReg-Inject™) and an existing product (chronOS Inject®) serving as a predicate device. A sheep model with a critical size drill hole in the tibial plateau was used. Holes of 8 mm diameter and 30 mm length were drilled bilaterally into the proximal area of the tibia and BoneReg-Inject™ or chronOS Inject® were injected into the right leg holes. Comparison of resorption and bone formation in vivo was made by X-ray micro-CT and histological evaluation after a live phase of 12 weeks. Long-term effects of BoneReg-Inject™ were studied using a 13-month live period. Significant differences were observed in (1) amount of new bone within implant (p < 0.001), higher in BoneReg-InjectTM, (2) signs of cartilage tissue (p = 0.003), more pronounced in BoneReg-InjectTM, and (3) signs of fibrous tissue (p < 0.001), less pronounced in BoneReg-InjectTM. Mineral content at 13 months postoperative was significantly higher than at 12 weeks (p < 0.001 and p < 0.05, for implant core and rim, respectively). The data demonstrate the potential of deacetylated chitin derivatives to stimulate bone formation.
Collapse
|
26
|
Raja IS, Preeth DR, Vedhanayagam M, Hyon SH, Lim D, Kim B, Rajalakshmi S, Han DW. Polyphenols-loaded electrospun nanofibers in bone tissue engineering and regeneration. Biomater Res 2021; 25:29. [PMID: 34563260 PMCID: PMC8466400 DOI: 10.1186/s40824-021-00229-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 08/30/2021] [Indexed: 01/27/2023] Open
Abstract
Bone is a complex structure with unique cellular and molecular process in its formation. Bone tissue regeneration is a well-organized and routine process at the cellular and molecular level in humans through the activation of biochemical pathways and protein expression. Though many forms of biomaterials have been applied for bone tissue regeneration, electrospun nanofibrous scaffolds have attracted more attention among researchers with their physicochemical properties such as tensile strength, porosity, and biocompatibility. When drugs, antibiotics, or functional nanoparticles are taken as additives to the nanofiber, its efficacy towards the application gets increased. Polyphenol is a versatile green/phytochemical small molecule playing a vital role in several biomedical applications, including bone tissue regeneration. When polyphenols are incorporated as additives to the nanofibrous scaffold, their combined properties enhance cell attachment, proliferation, and differentiation in bone tissue defect. The present review describes bone biology encompassing the composition and function of bone tissue cells and exemplifies the series of biological processes associated with bone tissue regeneration. We have highlighted the molecular mechanism of bioactive polyphenols involved in bone tissue regeneration and specified the advantage of electrospun nanofiber as a wound healing scaffold. As the polyphenols contribute to wound healing with their antioxidant and antimicrobial properties, we have compiled a list of polyphenols studied, thus far, for bone tissue regeneration along with their in vitro and in vivo experimental biological results and salient observations. Finally, we have elaborated on the importance of polyphenol-loaded electrospun nanofiber in bone tissue regeneration and discussed the possible challenges and future directions in this field.
Collapse
Affiliation(s)
| | - Desingh Raj Preeth
- Chemical Biology and Nanobiotechnology Laboratory, AU-KBC Research Centre, Anna University, MIT Campus, Chromepet, Chennai, 600 044, India
| | | | | | - Dohyung Lim
- Department of Mechanical Engineering, Sejong University, Seoul, 05006, South Korea
| | - Bongju Kim
- Dental Life Science Research Institute / Innovation Research & Support Center for Dental Science, Seoul National University Dental Hospital, Seoul, 03080, South Korea.
| | - Subramaniyam Rajalakshmi
- Chemical Biology and Nanobiotechnology Laboratory, AU-KBC Research Centre, Anna University, MIT Campus, Chromepet, Chennai, 600 044, India.
| | - Dong-Wook Han
- BIO-IT Fusion Technology Research Institute, Pusan National University, Busan, 46241, South Korea. .,Department of Cogno-Mechatronics Engineering, College of Nanoscience & Nanotechnology, Pusan National University, Busan, 46241, South Korea.
| |
Collapse
|