1
|
Mirsky NA, Ehlen QT, Greenfield JA, Antonietti M, Slavin BV, Nayak VV, Pelaez D, Tse DT, Witek L, Daunert S, Coelho PG. Three-Dimensional Bioprinting: A Comprehensive Review for Applications in Tissue Engineering and Regenerative Medicine. Bioengineering (Basel) 2024; 11:777. [PMID: 39199735 PMCID: PMC11351251 DOI: 10.3390/bioengineering11080777] [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: 06/18/2024] [Revised: 07/26/2024] [Accepted: 07/29/2024] [Indexed: 09/01/2024] Open
Abstract
Since three-dimensional (3D) bioprinting has emerged, it has continuously to evolved as a revolutionary technology in surgery, offering new paradigms for reconstructive and regenerative medical applications. This review highlights the integration of 3D printing, specifically bioprinting, across several surgical disciplines over the last five years. The methods employed encompass a review of recent literature focusing on innovations and applications of 3D-bioprinted tissues and/or organs. The findings reveal significant advances in the creation of complex, customized, multi-tissue constructs that mimic natural tissue characteristics, which are crucial for surgical interventions and patient-specific treatments. Despite the technological advances, the paper introduces and discusses several challenges that remain, such as the vascularization of bioprinted tissues, integration with the host tissue, and the long-term viability of bioprinted organs. The review concludes that while 3D bioprinting holds substantial promise for transforming surgical practices and enhancing patient outcomes, ongoing research, development, and a clear regulatory framework are essential to fully realize potential future clinical applications.
Collapse
Affiliation(s)
| | - Quinn T. Ehlen
- University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | | | | | - Blaire V. Slavin
- University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Vasudev Vivekanand Nayak
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Daniel Pelaez
- Dr. Nasser Ibrahim Al-Rashid Orbital Vision Research Center, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - David T. Tse
- Dr. Nasser Ibrahim Al-Rashid Orbital Vision Research Center, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Lukasz Witek
- Biomaterials Division, NYU Dentistry, New York, NY 10010, USA
- Department of Biomedical Engineering, New York University Tandon School of Engineering, Brooklyn, NY 11201, USA
- Hansjörg Wyss Department of Plastic Surgery, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Sylvia Daunert
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Paulo G. Coelho
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- DeWitt Daughtry Family Department of Surgery, Division of Plastic Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
2
|
Lei SS, Huang XW, Li LZ, Wang XP, Zhang Y, Li B, Shou D. Explorating the mechanism of Epimedii folium-Rhizoma drynariae herbal pair promoted bone defects healing through network pharmacology and experimental studies. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117329. [PMID: 37879510 DOI: 10.1016/j.jep.2023.117329] [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: 02/15/2023] [Revised: 10/05/2023] [Accepted: 10/16/2023] [Indexed: 10/27/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Bone defects are difficult to treat and have a high incidence of nonunion. The Epimedii folium-Rhizoma drynariae herbal pair (EDP) is a traditional Chinese medicine (TCM) used for treating bone diseases. However, the mechanisms by which EDP promotes osteogenesis or bone formation remain largely unclear. AIM OF THE STUDY This study aimed to investigate the mechanism of EDP promoted bone formation in bone defects using network pharmacology and experiments. MATERIALS AND METHODS The chemical components of EDP were analyzed by UHPLC-MS. The hub target and pathway enrichment analysis was conducted using molecular docking or network pharmacology. The pharmacological actions of EDP were determined by μCT and histopathology examination using a bone defect rat model. The effects of EDP on the mRNA expression of Bmp2, Smad2/5, Runx2, and Alp genes were measured by RT-PCR, while changes in the protein expressions of BMP2, COL1A1, SPP1, ALP, and RUNX2in the tibia tissues of the rats in response to EDP were analyzed by immunohistochemical staining or Western blot. We also performed cell viability assays, Alizarin Red and ALP staining assays, and RT-PCR to better understand how EDP affected osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs). RESULTS Identified 14 key compounds and 47 hub targets of EDP that may be involved in promoting osteogenesis to repair bone defects. And the BMP/Smad/Runx2 pathway was likely the key pathway through which EDP promoted bone defects repairing. The results of in vivo rat experiments indicated that EDP effectively promoted tibia repair in the model rats and activated the BMP/Smad/Runx2 pathway in the tibia tissue, with upregulating Bmp2, Bmpr1α, Smad2/5, Runx2, and Alp genes, and increased the protein expression of BMP2, COL1A1, RUNX2, and ALP. In vitro, EDP was found to increase the proliferation, differentiation, and mineralization in BMSCs- and also up-regulated the expression of key genes in the BMP/Smad/Runx2 pathway. CONCLUSION This study highlighted the ability of EDP to promote the osteogenic differentiation to enable bone repair by activating the BMP/Smad/Runx2 pathway.
Collapse
Affiliation(s)
- Shan Shan Lei
- Department of Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, Zhejiang, 310007, China
| | - Xiao Wen Huang
- Department of Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, Zhejiang, 310007, China
| | - Lin Zi Li
- Jingmen Central Hospital, 448000, Jingmen, China
| | - Xu Ping Wang
- Department of Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, Zhejiang, 310007, China
| | - Yang Zhang
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang 310053, China
| | - Bo Li
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang 310007, China.
| | - Dan Shou
- Department of Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, Zhejiang, 310007, China; School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 311402, China.
| |
Collapse
|
3
|
Gan J, Deng X, Le Y, Lai J, Liao X. The Development of Naringin for Use against Bone and Cartilage Disorders. Molecules 2023; 28:3716. [PMID: 37175126 PMCID: PMC10180405 DOI: 10.3390/molecules28093716] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 04/17/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
Bone and cartilage disorders are the leading causes of musculoskeletal disability. There is no absolute cure for all bone and cartilage disorders. The exploration of natural compounds for the potential therapeutic use against bone and cartilage disorders is proving promising. Among these natural chemicals, naringin, a flavanone glycoside, is a potential candidate due to its multifaceted pharmacological activities in bone and cartilage tissues. Emerging studies indicate that naringin may promote osteogenic differentiation, inhibit osteoclast formation, and exhibit protective effects against osteoporosis in vivo and in vitro. Many signaling pathways, such as BMP-2, Wnt/β-catenin, and VEGF/VEGFR, participate in the biological actions of naringin in mediating the pathological development of osteoporosis. In addition, the anti-inflammatory, anti-oxidative stress, and anti-apoptosis abilities of naringin also demonstrate its beneficial effects against bone and cartilage disorders, including intervertebral disc degeneration, osteoarthritis, rheumatoid arthritis, bone and cartilage tumors, and tibial dyschondroplasia. Naringin exhibits protective effects against bone and cartilage disorders. However, more efforts are still needed due to, at least in part, the uncertainty of drug targets. Further biological and pharmacological evaluations of naringin and its applications in bone tissue engineering, particularly its therapeutic effects against osteoporosis, might result in developing potential drug candidates.
Collapse
Affiliation(s)
- Juwen Gan
- Department of Pulmonary and Critical Care Medicine, Ganzhou People’s Hospital, Ganzhou 341000, China
| | - Xiaolan Deng
- Department of Pharmacy, Haikou Affiliated Hospital, Xiangya School of Medicine, Central South University, Haikou 570208, China
| | - Yonghong Le
- Department of Pulmonary and Critical Care Medicine, Ganzhou People’s Hospital, Ganzhou 341000, China
| | - Jun Lai
- Department of Pharmacy, Ganzhou People’s Hospital, Ganzhou 341000, China
| | - Xiaofei Liao
- Department of Pharmacy, Ganzhou People’s Hospital, Ganzhou 341000, China
| |
Collapse
|
4
|
Xiong Y, Mi BB, Lin Z, Hu YQ, Yu L, Zha KK, Panayi AC, Yu T, Chen L, Liu ZP, Patel A, Feng Q, Zhou SH, Liu GH. The role of the immune microenvironment in bone, cartilage, and soft tissue regeneration: from mechanism to therapeutic opportunity. Mil Med Res 2022; 9:65. [PMID: 36401295 PMCID: PMC9675067 DOI: 10.1186/s40779-022-00426-8] [Citation(s) in RCA: 88] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 10/30/2022] [Indexed: 11/21/2022] Open
Abstract
Bone, cartilage, and soft tissue regeneration is a complex spatiotemporal process recruiting a variety of cell types, whose activity and interplay must be precisely mediated for effective healing post-injury. Although extensive strides have been made in the understanding of the immune microenvironment processes governing bone, cartilage, and soft tissue regeneration, effective clinical translation of these mechanisms remains a challenge. Regulation of the immune microenvironment is increasingly becoming a favorable target for bone, cartilage, and soft tissue regeneration; therefore, an in-depth understanding of the communication between immune cells and functional tissue cells would be valuable. Herein, we review the regulatory role of the immune microenvironment in the promotion and maintenance of stem cell states in the context of bone, cartilage, and soft tissue repair and regeneration. We discuss the roles of various immune cell subsets in bone, cartilage, and soft tissue repair and regeneration processes and introduce novel strategies, for example, biomaterial-targeting of immune cell activity, aimed at regulating healing. Understanding the mechanisms of the crosstalk between the immune microenvironment and regeneration pathways may shed light on new therapeutic opportunities for enhancing bone, cartilage, and soft tissue regeneration through regulation of the immune microenvironment.
Collapse
Affiliation(s)
- Yuan Xiong
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Bo-Bin Mi
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Ze Lin
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Yi-Qiang Hu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China
| | - Le Yu
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, OH, 45701, USA
| | - Kang-Kang Zha
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China.,Key Laboratory of Biorheological Science and Technology,Ministry of Education College of Bioengineering, Chongqing University, Shapingba, Chongqing, 400044, China
| | - Adriana C Panayi
- Department of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02152, USA
| | - Tao Yu
- Department of Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Lang Chen
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China.,Department of Physics, Center for Hybrid Nanostructure (CHyN), University of Hamburg, Hamburg, 22761, Germany
| | - Zhen-Ping Liu
- Department of Physics, Center for Hybrid Nanostructure (CHyN), University of Hamburg, Hamburg, 22761, Germany.,Joint Laboratory of Optofluidic Technology and System,National Center for International Research on Green Optoelectronics, South China Academy of Advanced Optoelectronics, South China Normal University, Guangzhou, 510006, China
| | - Anish Patel
- Skeletal Biology Laboratory, Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02120, USA
| | - Qian Feng
- Key Laboratory of Biorheological Science and Technology,Ministry of Education College of Bioengineering, Chongqing University, Shapingba, Chongqing, 400044, China.
| | - Shuan-Hu Zhou
- Skeletal Biology Laboratory, Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02120, USA. .,Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA.
| | - Guo-Hui Liu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China. .,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, 430022, China.
| |
Collapse
|
5
|
Ye C, Chen J, Qu Y, Qi H, Wang Q, Yang Z, Wu A, Wang F, Li P. Naringin in the repair of knee cartilage injury via the TGF-β/ALK5/Smad2/3 signal transduction pathway combined with an acellular dermal matrix. J Orthop Translat 2022; 32:1-11. [PMID: 35591936 PMCID: PMC9072805 DOI: 10.1016/j.jot.2021.06.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 06/24/2021] [Accepted: 06/29/2021] [Indexed: 12/13/2022] Open
Affiliation(s)
- Chao Ye
- Orthopedics Department, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jing Chen
- Preventive Treatment of Disease Department, The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yi Qu
- Orthopedics Department, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Hui Qi
- Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Beijing, China
| | - Qingfu Wang
- Orthopedics Department, The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Zheng Yang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Aiming Wu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Fengxian Wang
- Orthopedics Department, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Pengyang Li
- Orthopedics Department, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Corresponding author. Orthopedics Department, Dongzhimen Hospital, Beijing University of Chinese Medicine, 5 Haiyuncang Street, Beijing, 100700, China.
| |
Collapse
|
6
|
Li Q, Liu S, Yang G, Li M, Qiao P, Xue Q. Naringenin inhibits autophagy and epithelial-mesenchymal transition of human lens epithelial cells by regulating the Smad2/3 pathway. Drug Dev Res 2021; 83:389-396. [PMID: 34402084 DOI: 10.1002/ddr.21868] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/03/2021] [Accepted: 08/05/2021] [Indexed: 12/15/2022]
Abstract
Cataract is the number one cause of blindness in the world. Fibrosis of the lens is the main cause of cataract. Pathological epithelial-mesenchymal transition (EMT) plays an important role in the development of fibrotic cataract. Inhibition of EMT may be an effective treatment for fibrosis of lens epithelial cells. Naringin (NRG) is one of the major citrus flavonoids, which has many pharmacological properties, including anti-inflammatory and cardioprotective. However, the effect of NRG on cataract induced by abnormal fibrosis of LECs is not clear. Herein, we found NRG inhibited transforming growth factor β2 (TGFβ2)-induced SRA01/04 cell viability. Additionally, NRG inhibited TGFβ2-induced cell migration and EMT. We further noticed that NRG inhibited autophagy and Smad2/3 phosphorylation in LECs. We therefore thought Naringenin inhibited autophagy and EMT of human LECs by regulating the Smad2/3 pathway. NRG could therefore serve as a promising drug for cataract treatment.
Collapse
Affiliation(s)
- Qingnan Li
- Department of Ophthalmology, Beijing Jishuitan Hospital, Beijing, China
| | - Shuang Liu
- Department of Ophthalmology, Beijing Jishuitan Hospital, Beijing, China
| | - Guang Yang
- Department of Ophthalmology, Beijing Jishuitan Hospital, Beijing, China
| | - Mingming Li
- Department of Ophthalmology, Beijing Friendship Hospital Affiliated to Capital Medical University, Beijing, China
| | - Peng Qiao
- Department of Ophthalmology, 3201 Hospital, Hanzhong, Shanxi, China
| | - Qiang Xue
- Department of Geriatrics, Beijing Jishuitan Hospital, Beijing, China
| |
Collapse
|
7
|
Ge X, Zhou G. Protective effects of naringin on glucocorticoid-induced osteoporosis through regulating the PI3K/Akt/mTOR signaling pathway. Am J Transl Res 2021; 13:6330-6341. [PMID: 34306372 PMCID: PMC8290725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 03/10/2021] [Indexed: 06/13/2023]
Abstract
OBJECTIVE To investigate the protective effects of Naringin on glucocorticoid-induced osteoporosis (GIOP) through the PI3K/AKT/mTOR signaling pathway in vivo and in vitro. METHODS Osteoblasts were cultured from the differentiated bone marrow mesenchymal stem cells (BM-MSCs) and were grouped as follows: the PBS group (the control group), the model group (Dexamethasone intervention), the LY294002 group (PI3K/AKT/mTOR pathway inhibitor intervention), the Naringin group (Naringin intervention), and the LY294002+ Naringin intervention group. Cell proliferation and differentiation were detected through cell counting kit-8 (CCK8) assay and alkaline phosphatase (ALP) staining, respectively. The formation of autophagosome was observed by Monodansylcadaverine (MDC) staining. Expressions of signaling pathway and autophagy related factors such as Beclin-1 and p62 were detected by qRT-PCR and western blot. Then, the rats were grouped as the PBS group (normal rats injected with PBS), the model group (GIOP rats injected with dexamethasone), the LY294002 group (GIOP rats injected with PI3K/AKT/mTOR pathway inhibitor LY294002), the Naringin group (GIOP rats injected with Naringin) and the LY294002+ Naringin group (GIOP rats injected with PI3K/AKT/mTOR pathway inhibitor LY294002 and Naringin). Bone mineral density and bone histomorphometry parameters of rats in each group were compared. In addition, the expressions of pathway and autophagy related factors in cartilage tissue of rats in each groups were also detected. RESULTS The proliferation and differentiation abilities of osteoblasts were increased with an increasing concentration of Naringin in a dose-dependent manner. Compared with the model group, the expression of PI3K/AKT/mTOR pathway related phosphorylated proteins, the proliferation and differentiation abilities of osteoblasts, the expression of autophagosome and autophagy related factors were all increased in the Naringin group, but contrary results were found in the LY294002 group (all P<0.05). In vivo, GIOP rats had improved bone mineral density and bone morphology parameters , and elevated expressions of autophagy related factors in cartilage tissue compared to the model group through Naringin intervention, while LY294002 intervention showed the opposite effects (all P<0.05). What is more, LY294002 partially reversed the effects of Naringin on osteogenic differentiation and bone morphological parameters in GIOP. CONCLUSION Naringin exerts protective effects in GIOP by the PI3K/AKT/mTOR pathway, which may be related to autophagy induction and enhanced proliferation of osteoblasts.
Collapse
Affiliation(s)
- Xingtao Ge
- Department of Orthopedics 4, Rizhao People’s HospitalRizhao, Shandong Province, China
| | - Gang Zhou
- Department of Orthopedic Surgery, Hainan General HospitalHaikou, Hainan Province, China
| |
Collapse
|