1
|
Lee JH, Shin SJ, Lee JH, Knowles JC, Lee HH, Kim HW. Adaptive immunity of materials: Implications for tissue healing and regeneration. Bioact Mater 2024; 41:499-522. [PMID: 39206299 PMCID: PMC11350271 DOI: 10.1016/j.bioactmat.2024.07.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/16/2024] [Accepted: 07/21/2024] [Indexed: 09/04/2024] Open
Abstract
Recent cumulative findings signify the adaptive immunity of materials as a key agenda in tissue healing that can improve regenerative events and outcomes. Modulating immune responses, mainly the recruitment and functions of T and B cells and their further interplay with innate immune cells (e.g., dendritic cells, macrophages) can be orchestrated by materials. For instance, decellularized matrices have been shown to promote muscle healing by inducing T helper 2 (Th2) cell immunity, while synthetic biopolymers exhibit differential effects on B cell responses and fibrosis compared decellularized matrices. We discuss the recent findings on how implantable materials instruct the adaptive immune events and the subsequent tissue healing process. In particular, we dissect the materials' physicochemical properties (shape, size, topology, degradation, rigidity, and matrix dynamic mechanics) to demonstrate the relations of these parameters with the adaptive immune responses in vitro and the underlying biological mechanisms. Furthermore, we present evidence of recent in vivo phenomena, including tissue healing, cancer progression, and fibrosis, wherein biomaterials potentially shape adaptive immune cell functions and in vivo outcomes. Our discussion will help understand the materials-regulated immunology events more deeply, and offer the design rationale of materials with tunable matrix properties for accelerated tissue repair and regeneration.
Collapse
Affiliation(s)
- Jung-Hwan Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea
- Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
- Cell & Matter Institute, Dankook University, Cheonan 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan 31116, Republic of Korea
| | - Seong-Jin Shin
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan 31116, Republic of Korea
| | - Jun Hee Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
- Cell & Matter Institute, Dankook University, Cheonan 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan 31116, Republic of Korea
| | - Jonathan C. Knowles
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan 31116, Republic of Korea
- UCL Eastman Dental Institute, University College London, London NW3 2PX, United Kingdom
| | - Hae-Hyoung Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea
- Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan 31116, Republic of Korea
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea
- Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
- Cell & Matter Institute, Dankook University, Cheonan 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan 31116, Republic of Korea
| |
Collapse
|
2
|
Shao YF, Wang H, Zhu Y, Peng Y, Bai F, Zhang J, Zhang KQ. Hydroxyapatite/Silk Fibroin Composite Scaffold with a Porous Structure and Mechanical Strength Similar to Cancellous Bone by Electric Field-Induced Gel Technology. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 39453828 DOI: 10.1021/acsami.4c12470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2024]
Abstract
Repair and regeneration of bone tissue defects is a multidimensional process that has been highly challenging to date. The artificial bone scaffold materials, which play a core role, still face the conflict that a biofriendly porous structure will reduce the mechanical performance and accelerate degradation. Herein, a multistage porous structured hydroxyapatite (HA)/silk fibroin (SF) composite scaffold (e-HA/SF) was successfully constructed by cleverly utilizing electric field-induced gel technology. The results indicated that the prepared e-HA/SF scaffolds possess biomimetic hierarchical porous structures with a suitable porosity similar to that of cancellous bone. The HA nanocrystals were uniformly encapsulated in the three-dimensional space of the composite scaffold, thus endowing the e-HA/SF composite scaffolds with an enhanced mechanical performance. Notably, the maximum compression stress and Young's modulus of e-HA/SF-2 scaffolds can reach 24.66 ± 0.88 and 28.91 ± 3.19 MPa, respectively, which are equivalent to those of cancellous bone. Such mechanical performance enhancement was previously unattainable through conventional freeze-drying strategies. Moreover, the introduction of bioactive nano-HA can trigger the optimal cell response in both static and dynamic cell culture experiments in vitro. The e-HA/SF composite scaffold developed in this study can better balance the conflict between the porous structure and mechanical and degradation properties of porous scaffolds.
Collapse
Affiliation(s)
- Yun-Fei Shao
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, P. R. China
| | - Hui Wang
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, P. R. China
| | - Yiran Zhu
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, P. R. China
| | - Yu Peng
- College of Advanced Material Engineering, Jiaxing Nanhu University, Jiaxing 314001, P. R. China
| | - Fengjiao Bai
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, P. R. China
| | - Jun Zhang
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, P. R. China
| | - Ke-Qin Zhang
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, P. R. China
| |
Collapse
|
3
|
Chundayil Kalathil N, Shah MR, Lailakumari VC, Prabhakaran P, Kumarapilla H, Kumar GSV. 3D Bilayered Hydrogel and Nanofiber Multifunctional Sponge Dressing: An Efficacious Healing Agent for Chronic Wound Healing. ACS APPLIED BIO MATERIALS 2024; 7:6492-6505. [PMID: 39271646 DOI: 10.1021/acsabm.4c00669] [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: 09/15/2024]
Abstract
Chronic wound management using biomaterial-based dressings has significantly impacted the standard and efficiency of wound healing. However, various available wound healing aids are ineffective in treating deep open injuries and chronic wounds such as diabetic wounds. Herein, we developed a 3D bilayered multifunctional sponge, which addresses the structural and functional issues faced by biomaterial dressings in treating deep and chronic wounds. The 3D bilayered sponge consists of a hydrogel base functionalized with wound healing peptide (Tylotoin)-carrying nanoparticles and topped with a nanofiber layer functionalized with an antimicrobial peptide (LLKKK18). The 3D bilayered sponge, with its highly porous, elastic, and enhanced fluid absorption ability, makes it a suitable wound treatment aid. The developed multifunctional 3D sponge shows antibacterial action and promotes a microenvironment similar to the extracellular matrix (ECM) in regulating dermal cell survival and migration. Study in a full-thickness skin defect diabetic mouse model has shown that the developed 3D bilayered sponge accelerated wound closure and promoted functional skin regeneration through reduced inflammation, faster granulation tissue formation, re-epithelialization, neovascularization, and skin appendage restoration, which make the developed 3D bilayered multifunctional sponge an efficient and advanced chronic wound management aid with potential for future clinical application.
Collapse
Affiliation(s)
- Nanditha Chundayil Kalathil
- Nano Drug Delivery Systems (NDDS), Rajiv Gandhi Centre for Biotechnology, Thycaud P.O, Thiruvananthapuram, Kerala 695014, India
- Research Centre, University of Kerala, Thiruvananthapuram, Kerala 695014, India
| | - Manan Rakesh Shah
- Nano Drug Delivery Systems (NDDS), Rajiv Gandhi Centre for Biotechnology, Thycaud P.O, Thiruvananthapuram, Kerala 695014, India
| | - Vipin Chandrasekharan Lailakumari
- Nano Drug Delivery Systems (NDDS), Rajiv Gandhi Centre for Biotechnology, Thycaud P.O, Thiruvananthapuram, Kerala 695014, India
- Regional Centre for Biotechnology (DBT-RCB), Faridabad, Haryana 121001, India
| | - Priya Prabhakaran
- Environmental Biology Laboratory, Rajiv Gandhi Centre for Biotechnology, Thycaud P.O, Poojappura, Thiruvananthapuram, Kerala 695014, India
| | - Harikrishnan Kumarapilla
- Environmental Biology Laboratory, Rajiv Gandhi Centre for Biotechnology, Thycaud P.O, Poojappura, Thiruvananthapuram, Kerala 695014, India
| | | |
Collapse
|
4
|
Arab S, Bahraminasab M, Asgharzade S, Doostmohammadi A, Zadeh ZK, Nooshabadi VT. On the osteogenic differentiation of dental pulp stem cells by a fabricated porous nano-hydroxyapatite substrate loaded with sodium fluoride. BMC Oral Health 2024; 24:1218. [PMID: 39402484 PMCID: PMC11476061 DOI: 10.1186/s12903-024-04987-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 10/01/2024] [Indexed: 10/19/2024] Open
Abstract
In the present study, nano-hydroxyapatite (n-HA) powder was extracted from carp bone waste to fabricate porous n-HA substrates by a molding and sintering process. Subsequently, the substrates were loaded with different amounts of sodium fluoride (NaF) through immersion in NaF suspensions for 10, 7.5, and 5 min. The NaF-loaded n-HA substrates were then examined for their structural and physical properties, chemical bonds, loading and release profile, pH changes, cytotoxicity, and osteogenic effect on dental pulp stem cells (DPSCs) at the level of RNA and protein expression. The results showed that the n-HA substrates were porous (> 40% porosity) and had rough surfaces. The NaF could be successfully loaded on the substrates, which was 6.43, 4.50, and 1.47 mg, respectively for n-HA substrates with immersion times of 10, 7.5, and 5 min in the NaF suspensions. It was observed that the NaF release rate was rather fast during the first 24 h in all groups (39.06%, 36.43%, and 39.57% for 10, 7.5, and 5 min, respectively), and decreased dramatically after that, indicating a slow detachment of NaF. Furthermore, the pH of the medium related to all materials was changed during the first 4 days of immersion (from 7.38 to pH of about 7.85, 7.84, 7.63, and 7.66 for C0, C5, C7.5, and C10, respectively). The pH of media associated with the C7.5, and C10 increased up to 4 days and remained relatively constant until day 14 (pH = 7.6). The results of the cytotoxicity assay rejected any toxicity of the fabricated NaF-loaded n-HA substrates on DPSCs, and the cells could adhere to their surfaces with enlarged morphology. The results showed no effect on the osteogenic differentiation at the protein level. Nevertheless, this effect was observed at the gene level.
Collapse
Affiliation(s)
- Samaneh Arab
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Marjan Bahraminasab
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran.
| | - Samira Asgharzade
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
- Department of Molecular Medicine, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Ali Doostmohammadi
- Student Research Committee, Semnan University of Medical Sciences, Semnan, Iran
| | - Zahra Khatib Zadeh
- Student Research Committee, Semnan University of Medical Sciences, Semnan, Iran
| | - Vajihe Taghdiri Nooshabadi
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
5
|
Han EH, Cho SH, Lee SN, Cho MY, Lee H, Lee SY, Ngoc Thi Tran C, Park HS, Min JY, Kim HM, Park MS, Kim TD, Lim YT, Hong KS. 3D Scaffold-Based Culture System Enhances Preclinical Evaluation of Natural Killer Cell Therapy in A549 Lung Cancer Cells. ACS APPLIED BIO MATERIALS 2024. [PMID: 39392900 DOI: 10.1021/acsabm.4c00800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2024]
Abstract
Cell-based immunotherapies have emerged as promising cancer treatment modalities, demonstrating remarkable clinical efficacy. As interest in applying immune cell-based therapies to solid tumors has gained momentum, experimental models that enable long-term monitoring and mimic clinical administration are increasingly necessary. This study explores the potential of scaffold-based cell culture technologies, specifically three-dimensional (3D) extracellular matrix (ECM)-like frameworks, as promising solutions. These frameworks facilitate unhindered immune cell growth and enable continuous cancer cell culture. The three-dimensional (3D) cell culture model was developed using tailored scaffolds for natural killer (NK) cell culture. Within this framework, A549 lung cancer cells were cocultured with NK cells, allowing real-time monitoring for up to 28 days. The expression of critical markers associated with anticancer drug resistance and epithelial-mesenchymal transition (EMT) was evaluated in cancer cells within this 3D culture context. Compared to conventional 2D monolayer cultures, this 3D scaffold-based culture revealed that solid tumor cells, specifically A549 cells, exhibited heightened resistance to anticancer drugs. Additionally, the 3D culture environment upregulated the expression of EMT markers namely vimentin, N-cadherin, and fibronectin, while NK and zEGFR-CAR-NK cells displayed anticancer effects. In the two-dimensional (2D) coculture, only zEGFR-CAR-NK cells exhibited such effects in the 3D coculture system, highlighting an intriguing inconsistency with the 2D culture model, further confirmed by in vivo experiments. This in vitro 3D cell culture model reliably predicts outcomes in NK immunotherapy experiments. Thus, it represents a valuable tool for investigating drug resistance mechanisms and assessing the efficacy of immune cell-based therapies. By bridging the gap between in vitro and in vivo investigations, this model effectively translates potential treatments into animal models and facilitates rigorous preclinical evaluations.
Collapse
Affiliation(s)
- Eun Hee Han
- Biopharmaceutical Research Center, Ochang Institute of Biological and Environmental Science, Korea Basic Science Institute (KBSI), Cheongju 28119, Republic of Korea
- Korea University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Sun-Hee Cho
- Biopharmaceutical Research Center, Ochang Institute of Biological and Environmental Science, Korea Basic Science Institute (KBSI), Cheongju 28119, Republic of Korea
| | - Sang Nam Lee
- SKKU Advanced Institute of Nanotechnology (SAINT) and School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Mi Young Cho
- Biopharmaceutical Research Center, Ochang Institute of Biological and Environmental Science, Korea Basic Science Institute (KBSI), Cheongju 28119, Republic of Korea
- SKKU Advanced Institute of Nanotechnology (SAINT) and School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Hyunseung Lee
- Biopharmaceutical Research Center, Ochang Institute of Biological and Environmental Science, Korea Basic Science Institute (KBSI), Cheongju 28119, Republic of Korea
| | - Soo Yun Lee
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Chau Ngoc Thi Tran
- Biopharmaceutical Research Center, Ochang Institute of Biological and Environmental Science, Korea Basic Science Institute (KBSI), Cheongju 28119, Republic of Korea
- Department of Chemistry, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Hye Sun Park
- Biopharmaceutical Research Center, Ochang Institute of Biological and Environmental Science, Korea Basic Science Institute (KBSI), Cheongju 28119, Republic of Korea
| | - Jin Young Min
- Biopharmaceutical Research Center, Ochang Institute of Biological and Environmental Science, Korea Basic Science Institute (KBSI), Cheongju 28119, Republic of Korea
| | - Hye Min Kim
- Biopharmaceutical Research Center, Ochang Institute of Biological and Environmental Science, Korea Basic Science Institute (KBSI), Cheongju 28119, Republic of Korea
- Korea University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Min Sung Park
- Biopharmaceutical Research Center, Ochang Institute of Biological and Environmental Science, Korea Basic Science Institute (KBSI), Cheongju 28119, Republic of Korea
- Korea University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Tae-Don Kim
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Yong Taik Lim
- SKKU Advanced Institute of Nanotechnology (SAINT) and School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Kwan Soo Hong
- Biopharmaceutical Research Center, Ochang Institute of Biological and Environmental Science, Korea Basic Science Institute (KBSI), Cheongju 28119, Republic of Korea
- Department of Chemistry, Chung-Ang University, Seoul 06974, Republic of Korea
| |
Collapse
|
6
|
Dan X, Li S, Chen H, Xue P, Liu B, Ju Y, Lei L, Li Y, Fan X. Tailoring biomaterials for skin anti-aging. Mater Today Bio 2024; 28:101210. [PMID: 39285945 PMCID: PMC11402947 DOI: 10.1016/j.mtbio.2024.101210] [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: 06/07/2024] [Revised: 08/19/2024] [Accepted: 08/23/2024] [Indexed: 09/19/2024] Open
Abstract
Skin aging is the phenomenon of degenerative changes in the structure and function of skin tissues over time and is manifested by a gradual loss of skin elasticity and firmness, an increased number of wrinkles, and hyperpigmentation. Skin anti-aging refers to a reduction in the skin aging phenomenon through medical cosmetic technologies. In recent years, new biomaterials have been continuously developed for improving the appearance of the skin through mechanical tissue filling, regulating collagen synthesis and degradation, inhibiting pigmentation, and repairing the skin barrier. This review summarizes the mechanisms associated with skin aging, describes the biomaterials that are commonly used in medical aesthetics and their possible modes of action, and discusses the application strategies of biomaterials in this area. Moreover, the synergistic effects of such biomaterials and other active ingredients, such as stem cells, exosomes, growth factors, and antioxidants, on tissue regeneration and anti-aging are evaluated. Finally, the possible challenges and development prospects of biomaterials in the field of anti-aging are discussed, and novel ideas for future innovations in this area are summarized.
Collapse
Affiliation(s)
- Xin Dan
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Songjie Li
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Han Chen
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Ping Xue
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Bo Liu
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Yikun Ju
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Lanjie Lei
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, 310015, China
| | - Yang Li
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Xing Fan
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| |
Collapse
|
7
|
Haider A, Khan S, Iqbal DN, Khan SU, Haider S, Mohammad K, Mustfa G, Rizwan M, Haider A. Chitosan as a tool for tissue engineering and rehabilitation: Recent developments and future perspectives - A review. Int J Biol Macromol 2024; 278:134172. [PMID: 39111484 DOI: 10.1016/j.ijbiomac.2024.134172] [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: 05/05/2024] [Revised: 07/17/2024] [Accepted: 07/24/2024] [Indexed: 08/17/2024]
Abstract
Chitosan has established itself as a multifunctional and auspicious biomaterial within the domain of tissue engineering, presenting a decade of uninterrupted advancements and novel implementations. This article provides a comprehensive overview of the most recent developments in chitosan-based tissue engineering, focusing on significant progress made in the last ten years. An exploration is conducted of the various techniques utilized in the modification of chitosan and the production of scaffolds, with an analysis of their effects on cellular reactions and tissue regeneration. The investigation focuses on the integration of chitosan with other biomaterials and the addition of bioactive agents to improve their functionalities. Upon careful analysis of the in vitro and in vivo research, it becomes evident that chitosan effectively stimulates cell adhesion, proliferation, and differentiation. Furthermore, we offer valuable perspectives on the dynamic realm of chitosan-based approaches tailored to distinct tissue categories, including nerve, bone, cartilage, and skin. The review concludes with a discussion of prospective developments, with particular attention given to possible directions for additional study, translational implementations, and the utilization of chitosan to tackle existing obstacles in the field of tissue engineering. This extensive examination provides a significant amalgamation of the advancements achieved over the previous decade and directs scholars towards uncharted territories in chitosan-based tissue engineering.
Collapse
Affiliation(s)
- Ammar Haider
- Department of Chemistry, The University of Lahore, Lahore 54000, Pakistan
| | - Shabana Khan
- Department of Chemistry, The University of Lahore, Lahore 54000, Pakistan
| | - Dure Najaf Iqbal
- Department of Chemistry, The University of Lahore, Lahore 54000, Pakistan.
| | - Salah Uddin Khan
- Sustainable Energy Technologies Center, College of Engineering, King Saud University, P.O. Box 800, Riyadh 11421, Saudi Arabia; King Salman Center for Disability Research, Riyadh 11614, Saudi Arabia.
| | - Sajjad Haider
- Chemical Engineering Department, College of Engineering, King Saud University, P.O. Box 800, Riyadh 11421, Saudi Arabia
| | - Khaled Mohammad
- Chemical Engineering Department, College of Engineering, King Saud University, P.O. Box 800, Riyadh 11421, Saudi Arabia
| | - Ghulam Mustfa
- Department of Chemistry, The University of Lahore, Lahore 54000, Pakistan
| | - Muhammad Rizwan
- Department of Chemistry, The University of Lahore, Lahore 54000, Pakistan
| | - Adnan Haider
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi 46000, Pakistan
| |
Collapse
|
8
|
Lu H, Li Z, Duan Z, Liao Y, Liu K, Zhang Y, Fan L, Xu T, Yang D, Wang S, Fu Y, Xiang H, Chen Y, Li G. Photothermal Catalytic Reduction and Bone Tissue Engineering Towards a Three-in-One Therapy Strategy for Osteosarcoma. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2408016. [PMID: 39165073 DOI: 10.1002/adma.202408016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/15/2024] [Indexed: 08/22/2024]
Abstract
Osteosarcoma is one of the most dreadful bone neoplasms in young people, necessitating the development of innovative therapies that can effectively eliminate tumors while minimizing damage to limb function. An ideal therapeutic strategy should possess three essential capabilities: antitumor effects, tissue-protective properties, and the ability to enhance osteogenesis. In this study, self-assembled Ce-substituted molybdenum blue (CMB) nanowheel crystals are synthesized and loaded onto 3D-printed bioactive glass (CMB@BG) scaffolds to develop a unique three-in-one treatment approach for osteosarcoma. The CMB@BG scaffolds exhibit outstanding photothermally derived tumor ablation within the near-infrared-II window due to the surface plasmon resonance properties of the CMB nanowheel crystals. Furthermore, the photothermally synergistic catalytic effect of CMB promotes the rapid scavenging of reactive oxygen species caused by excessive heat, thereby suppressing inflammation and protecting surrounding tissues. The CMB@BG scaffolds possess pro-proliferation and pro-differentiation capabilities that efficiently accelerate bone regeneration within bone defects. Altogether, the CMB@BG scaffolds that combine highly efficient tumor ablation, tissue protection based on anti-inflammatory mechanisms, and enhanced osteogenic ability are likely to be a point-to-point solution for the comprehensive therapeutic needs of osteosarcoma.
Collapse
Affiliation(s)
- Hengli Lu
- Department of Orthopaedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Zihua Li
- Department of Orthopaedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Zhengwei Duan
- Department of Orthopaedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Yuxin Liao
- Department of Orthopaedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Kaiyuan Liu
- Department of Bone Tumor Surgery, Shanghai General Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 200025, P. R. China
| | - Yiwei Zhang
- Department of Orthopaedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Lin Fan
- Department of Orthopaedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Tianyang Xu
- Department of Orthopaedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Dong Yang
- Department of Orthopaedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Sen Wang
- Department of Orthopaedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Yuesong Fu
- Department of Orthopaedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Huijing Xiang
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute of Shanghai University, Wenzhou, Zhejiang, 325088, P. R. China
- Shanghai Institute of Materdicine, Shanghai, 200051, P. R. China
| | - Guodong Li
- Department of Orthopaedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| |
Collapse
|
9
|
Mikaeeli Kangarshahi B, Naghib SM, Rabiee N. 3D printing and computer-aided design techniques for drug delivery scaffolds in tissue engineering. Expert Opin Drug Deliv 2024:1-22. [PMID: 39323396 DOI: 10.1080/17425247.2024.2409913] [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: 01/20/2024] [Revised: 09/19/2024] [Accepted: 09/24/2024] [Indexed: 09/27/2024]
Abstract
INTRODUCTION The challenge in tissue engineering lies in replicating the intricate structure of the native extracellular matrix. Recent advancements in AM, notably 3D printing, offer unprecedented capabilities to tailor scaffolds precisely, controlling properties like structure and bioactivity. CAD tools complement this by facilitating design using patient-specific data. AREA’S COVERED This review introduces additive manufacturing (AM) and computer-aided design (CAD) as pivotal tools in advancing tissue engineering, particularly cartilage regeneration. This article explores various materials utilized in AM, focusing on polymers and hydrogels for their advantageous properties in tissue engineering applications. Integrating bioactive molecules, including growth factors, into scaffolds to promote tissue regeneration is discussed alongside strategies involving different cell sources, such as stem cells, to enhance tissue development within scaffold matrices. EXPERT OPINION Applications of AM and CAD in addressing specific challenges like osteochondral defects and osteoarthritis in cartilage tissue engineering are highlighted. This review consolidates current research findings, offering expert insights into the evolving landscape of AM and CAD technologies in advancing tissue engineering, particularly in cartilage regeneration.
Collapse
Affiliation(s)
- Babak Mikaeeli Kangarshahi
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology, Tehran, Iran
| | - Seyed Morteza Naghib
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology, Tehran, Iran
| | - Navid Rabiee
- Department of Biomaterials, Saveetha Dental College and Hospitals, SIMATS, Saveetha University, Chennai, India
| |
Collapse
|
10
|
Agarwal P, Mathur V, Kasturi M, Srinivasan V, Seetharam RN, S Vasanthan K. A Futuristic Development in 3D Printing Technique Using Nanomaterials with a Step Toward 4D Printing. ACS OMEGA 2024; 9:37445-37458. [PMID: 39281933 PMCID: PMC11391532 DOI: 10.1021/acsomega.4c04123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/27/2024] [Accepted: 08/06/2024] [Indexed: 09/18/2024]
Abstract
3D bioprinting has shown great promise in tissue engineering and regenerative medicine for creating patient-specific tissue scaffolds and medicinal devices. The quickness, accurate imaging, and design targeting of this emerging technology have excited biomedical engineers and translational medicine researchers. Recently, scaffolds made from 3D bioprinted tissue have become more clinically effective due to nanomaterials and nanotechnology. Because of quantum confinement effects and high surface area/volume ratios, nanomaterials and nanotechnological techniques have unique physical, chemical, and biological features. The use of nanomaterials and 3D bioprinting has led to scaffolds with improved physicochemical and biological properties. Nanotechnology and nanomaterials affect 3D bioprinted tissue engineered scaffolds for regenerative medicine and tissue engineering. Biomaterials and cells that respond to stimuli change the structural shape in 4D bioprinting. With such dynamic designs, tissue architecture can change morphologically. New 4D bioprinting techniques will aid in bioactuation, biorobotics, and biosensing. The potential of 4D bioprinting in biomedical technologies is also discussed in this article.
Collapse
Affiliation(s)
- Prachi Agarwal
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Karnataka, Manipal 576104, India
| | - Vidhi Mathur
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Karnataka, Manipal 576104, India
| | - Meghana Kasturi
- Department of Mechanical Engineering, University of Michigan, Dearborn, Michigan 48128, United States
| | - Varadharajan Srinivasan
- Manipal Institute of Technology, Manipal Academy of Higher Education, Karnataka, Manipal 576104, India
| | - Raviraja N Seetharam
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Karnataka, Manipal 576104, India
| | - Kirthanashri S Vasanthan
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Karnataka, Manipal 576104, India
| |
Collapse
|
11
|
Culbreath CJ, McCullen SD, Mefford OT. Controlling Mechanical Properties of Medical-Grade Scaffolds through Electrospinning Parameter Selection. ACS OMEGA 2024; 9:36982-36992. [PMID: 39246470 PMCID: PMC11375708 DOI: 10.1021/acsomega.4c01864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 07/28/2024] [Accepted: 07/30/2024] [Indexed: 09/10/2024]
Abstract
Electrospinning (ES) is a versatile process mode for creating fibrous materials with various structures that have broad applications ranging from regenerative medicine to tissue engineering and surgical mesh implants. The recent commercialization of this technology for implant use has driven the use of resorbable electrospun products. Resorbable electrospun meshes offer great promise as temporary implants that can utilize the layer upon layer method of additive manufacturing to incorporate porosity as a function of process parameters into a scaffold structure. The interconnected porosity and feature size known to ES have previously been observed to hold great potential for simulating the natural cellular environment of soft tissue. This microstructure, proper degradation kinetics, and mechanical properties combine to provide the design basis for artificial tissue structures that could aid in not only wound healing but also true tissue engineering and regenerative medicine. While current advancement in the field is understood to be limited by material properties, the importance of optimizing mechanical properties with currently available materials should not be overlooked. This work investigated the process parameter effects and interactions that control the structure-property relationship for a range of medical-grade aliphatic polyester materials with a range of intrinsic properties. An ε-caprolactone homopolymer (PCL), l-lactide homopolymer (PLLA), and Lactoflex, a copolymer with intermediate properties relative to the homopolymers, were characterized before, during, and after the additive manufacturing process. The interacting effects of process parameters, distance to collector, and dispensing rate were shown to produce variable-density, nonwoven scaffold structures. The resorbable mesh scaffolds of PLLA, PCL, and Lactoflex demonstrated a broad range of mechanical properties (approximately 1-10 MPa ultimate tensile strength and 5-390 MPa tensile modulus). Postprocessing of scaffolds demonstrated removal of solvents and preservation of micrometer-sized features. Resorbable polymers and electrospinning can produce scaffold materials with excellent features and offer tremendous potential in the field of implantable resorbable devices.
Collapse
Affiliation(s)
- Clayton J Culbreath
- Poly-Med, Inc. Anderson, South Carolina 29625, United States
- Department of Materials Science and Engineering, Clemson University, Clemson, South Carolina 29634, United States
| | - Seth D McCullen
- Poly-Med, Inc. Anderson, South Carolina 29625, United States
- Department of Materials Science and Engineering, Clemson University, Clemson, South Carolina 29634, United States
| | - O Thompson Mefford
- Department of Materials Science and Engineering, Clemson University, Clemson, South Carolina 29634, United States
- Department of Bioengineering, Clemson University, Clemson, South Carolina 29634, United States
| |
Collapse
|
12
|
Cai G, Ren L, Yu J, Jiang S, Liu G, Wu S, Cheng B, Li W, Xia J. A Microenvironment-Responsive, Controlled Release Hydrogel Delivering Embelin to Promote Bone Repair of Periodontitis via Anti-Infection and Osteo-Immune Modulation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403786. [PMID: 38978324 PMCID: PMC11425865 DOI: 10.1002/advs.202403786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/12/2024] [Indexed: 07/10/2024]
Abstract
Periodontitis, a prevalent chronic inflammatory disease, poses significant challenges for effective treatment due to its complex etiology involving specific bacteria and the inflammatory immune microenvironment. Here, this study presents a novel approach for the targeted treatment of periodontitis utilizing the immunomodulatory and antibacterial properties of Embelin, a plant-derived compound, within an injectable hydrogel system. The developed Carboxymethyl Chitosan-Oxidized Dextran (CMCS-OD) hydrogel formed via dynamic chemical bonds exhibited self-healing capabilities and pH-responsive behavior, thereby facilitating the controlled release of Embelin and enhancing its efficacy in a dynamic oral periodontitis microenvironment. This study demonstrates that this hydrogel system effectively prevents bacterial invasion and mitigates excessive immune response activation. Moreover, it precisely modulates macrophage M1/M2 phenotypes and suppresses inflammatory cytokine expression, thereby fostering a conducive environment for bone regeneration and addressing periodontitis-induced bone loss. These findings highlight the potential of the approach as a promising strategy for the clinical management of periodontitis-induced bone destruction.
Collapse
Affiliation(s)
- Guanming Cai
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, P. R. China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China
| | - Lin Ren
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, P. R. China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China
| | - Jiali Yu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, P. R. China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China
| | - Siqi Jiang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, P. R. China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China
| | - Gen Liu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, P. R. China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China
| | - Shujie Wu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, P. R. China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China
| | - Bin Cheng
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, P. R. China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China
| | - Weichang Li
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, P. R. China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China
| | - Juan Xia
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, P. R. China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China
| |
Collapse
|
13
|
Callegari F, Brofiga M, Tedesco M, Massobrio P. Electrophysiological features of cortical 3D networks are deeply modulated by scaffold properties. APL Bioeng 2024; 8:036112. [PMID: 39193551 PMCID: PMC11348497 DOI: 10.1063/5.0214745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 08/05/2024] [Indexed: 08/29/2024] Open
Abstract
Three-dimensionality (3D) was proven essential for developing reliable models for different anatomical compartments and many diseases. However, the neuronal compartment still poses a great challenge as we still do not understand precisely how the brain computes information and how the complex chain of neuronal events can generate conscious behavior. Therefore, a comprehensive model of neuronal tissue has not yet been found. The present work was conceived in this framework: we aimed to contribute to what must be a collective effort by filling in some information on possible 3D strategies to pursue. We compared directly different kinds of scaffolds (i.e., PDMS sponges, thermally crosslinked hydrogels, and glass microbeads) in their effect on neuronal network activity recorded using micro-electrode arrays. While the overall rate of spiking activity remained consistent, the type of scaffold had a notable impact on bursting dynamics. The frequency, density of bursts, and occurrence of random spikes were all affected. The examination of inter-burst intervals revealed distinct burst generation patterns unique to different scaffold types. Network burst propagation unveiled divergent trends among configurations. Notably, it showed the most differences, underlying that functional variations may arise from a different 3D spatial organization. This evidence suggests that not all 3D neuronal constructs can sustain the same level of richness of activity. Furthermore, we commented on the reproducibility, efficacy, and scalability of the methods, where the beads still offer superior performances. By comparing different 3D scaffolds, our results move toward understanding the best strategies to develop functional 3D neuronal units for reliable pre-clinical studies.
Collapse
Affiliation(s)
- Francesca Callegari
- Department of Informatics, Bioengineering, Robotics, and Systems Engineering (DIBRIS), University of Genova, Genova, Italy
| | | | - Mariateresa Tedesco
- Department of Informatics, Bioengineering, Robotics, and Systems Engineering (DIBRIS), University of Genova, Genova, Italy
| | | |
Collapse
|
14
|
Todd L, Chin MHW, Coppens MO. Two conjectures on 3D Voronoi structures: a toolkit with biomedical case studies. MOLECULAR SYSTEMS DESIGN & ENGINEERING 2024; 9:912-919. [PMID: 39205672 PMCID: PMC11348831 DOI: 10.1039/d4me00036f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 05/19/2024] [Indexed: 09/04/2024]
Abstract
3D Voronoi scaffolds are widely applied in the field of additive manufacturing as they are known for their light weight structural resilience and share many topological similarities to various natural (bone, tumours, lymph node) and synthetic environments (foam, functionally gradient porous materials). Unfortunately, the structural design features that promote these topological similarities (such as the number of vertices) are often unpredictable and require the trial and error of varying design features to achieve the desired 3D Voronoi structure. This article provides a toolkit, consisting of equations, based on over 12 000 3D Voronoi structures. These equations allow design features, such as the number of generating points (G), to be efficiently and accurately predicted based on the desired structural parameters (within ±3G). Based on these equations we are proposing, to the best of our knowledge, two new mathematical conjectures that relate the number of vertices or edges, and the average edge length to G in Voronoi structures. These equations have been validated for a wide range of parameter values and Voronoi network sizes. A design code is provided allowing any of over 12 000 structures to be selected, easily adjusted based on user requirements, and 3D printed. Biomedical case studies relevant to T-cell culturing, bone scaffolds and kidney tumours are presented to illustrate the design code.
Collapse
Affiliation(s)
- Lucy Todd
- Centre for Nature Inspired Engineering & Department of Chemical Engineering, University College London Torrington Place London WC1E 7JE United Kingdom
| | - Matthew H W Chin
- Centre for Nature Inspired Engineering & Department of Chemical Engineering, University College London Torrington Place London WC1E 7JE United Kingdom
| | - Marc-Olivier Coppens
- Centre for Nature Inspired Engineering & Department of Chemical Engineering, University College London Torrington Place London WC1E 7JE United Kingdom
| |
Collapse
|
15
|
Behroozi Kohlan T, Wen Y, Mini C, Finne-Wistrand A. Schiff base crosslinked hyaluronic acid hydrogels with tunable and cell instructive time-dependent mechanical properties. Carbohydr Polym 2024; 338:122173. [PMID: 38763720 DOI: 10.1016/j.carbpol.2024.122173] [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: 02/17/2024] [Revised: 03/30/2024] [Accepted: 04/16/2024] [Indexed: 05/21/2024]
Abstract
The dynamic interplay between cells and their native extracellular matrix (ECM) influences cellular behavior, imposing a challenge in biomaterial design. Dynamic covalent hydrogels are viscoelastic and show self-healing ability, making them a potential scaffold for recapitulating native ECM properties. We aimed to implement kinetically and thermodynamically distinct crosslinkers to prepare self-healing dynamic hydrogels to explore the arising properties and their effects on cellular behavior. To do so, aldehyde-substituted hyaluronic acid (HA) was synthesized to generate imine, hydrazone, and oxime crosslinked dynamic covalent hydrogels. Differences in equilibrium constants of these bonds yielded distinct properties including stiffness, stress relaxation, and self-healing ability. The effects of degree of substitution (DS), polymer concentration, crosslinker to aldehyde ratio, and crosslinker functionality on hydrogel properties were evaluated. The self-healing ability of hydrogels was investigated on samples of the same and different crosslinkers and DS to obtain hydrogels with gradient properties. Subsequently, human dermal fibroblasts were cultured in 2D and 3D to assess the cellular response considering the dynamic properties of the hydrogels. Moreover, assessing cell spreading and morphology on hydrogels having similar modulus but different stress relaxation rates showed the effects of matrix viscoelasticity with higher cell spreading in slower relaxing hydrogels.
Collapse
Affiliation(s)
- Taha Behroozi Kohlan
- Department of Fibre and Polymer Technology, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Teknikringen, 56-58, SE 10044 Stockholm, Sweden
| | - Yanru Wen
- Department of Fibre and Polymer Technology, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Teknikringen, 56-58, SE 10044 Stockholm, Sweden
| | - Carina Mini
- Department of Fibre and Polymer Technology, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Teknikringen, 56-58, SE 10044 Stockholm, Sweden
| | - Anna Finne-Wistrand
- Department of Fibre and Polymer Technology, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Teknikringen, 56-58, SE 10044 Stockholm, Sweden.
| |
Collapse
|
16
|
Abed H, Radha R, Anjum S, Paul V, AlSawaftah N, Pitt WG, Ashammakhi N, Husseini GA. Targeted Cancer Therapy-on-A-Chip. Adv Healthc Mater 2024:e2400833. [PMID: 39101627 DOI: 10.1002/adhm.202400833] [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: 03/04/2024] [Revised: 06/15/2024] [Indexed: 08/06/2024]
Abstract
Targeted cancer therapy (TCT) is gaining increased interest because it reduces the risks of adverse side effects by specifically treating tumor cells. TCT testing has traditionally been performed using two-dimensional (2D) cell culture and animal studies. Organ-on-a-chip (OoC) platforms have been developed to recapitulate cancer in vitro, as cancer-on-a-chip (CoC), and used for chemotherapeutics development and testing. This review explores the use of CoCs to both develop and test TCTs, with a focus on three main aspects, the use of CoCs to identify target biomarkers for TCT development, the use of CoCs to test free, un-encapsulated TCTs, and the use of CoCs to test encapsulated TCTs. Despite current challenges such as system scaling, and testing externally triggered TCTs, TCToC shows a promising future to serve as a supportive, pre-clinical platform to expedite TCT development and bench-to-bedside translation.
Collapse
Affiliation(s)
- Heba Abed
- Department of Chemical and Biological Engineering, American University of Sharjah, Sharjah, UAE
| | - Remya Radha
- Department of Chemical and Biological Engineering, American University of Sharjah, Sharjah, UAE
| | - Shabana Anjum
- Department of Chemical and Biological Engineering, American University of Sharjah, Sharjah, UAE
| | - Vinod Paul
- Materials Science and Engineering PhD program, College of Arts and Sciences, American University of Sharjah, Sharjah, UAE
| | - Nour AlSawaftah
- Materials Science and Engineering PhD program, College of Arts and Sciences, American University of Sharjah, Sharjah, UAE
| | - William G Pitt
- Department of Chemical Engineering, Brigham Young University, Provo, UT, 84602, USA
| | - Nureddin Ashammakhi
- Institute for Quantitative Health Science and Engineering (IQ) and Department of Biomedical Engineering (BME), Michigan State University, East Lansing, MI, 48824, USA
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095-1600, USA
| | - Ghaleb A Husseini
- Department of Chemical and Biological Engineering, American University of Sharjah, Sharjah, UAE
- Materials Science and Engineering PhD program, College of Arts and Sciences, American University of Sharjah, Sharjah, UAE
| |
Collapse
|
17
|
Di Berardino C, Peserico A, Camerano Spelta Rapini C, Liverani L, Capacchietti G, Russo V, Berardinelli P, Unalan I, Damian-Buda AI, Boccaccini AR, Barboni B. Bioengineered 3D ovarian model for long-term multiple development of preantral follicle: bridging the gap for poly(ε-caprolactone) (PCL)-based scaffold reproductive applications. Reprod Biol Endocrinol 2024; 22:95. [PMID: 39095895 PMCID: PMC11295475 DOI: 10.1186/s12958-024-01266-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 07/24/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Assisted Reproductive Technologies (ARTs) have been validated in human and animal to solve reproductive problems such as infertility, aging, genetic selection/amplification and diseases. The persistent gap in ART biomedical applications lies in recapitulating the early stage of ovarian folliculogenesis, thus providing protocols to drive the large reserve of immature follicles towards the gonadotropin-dependent phase. Tissue engineering is becoming a concrete solution to potentially recapitulate ovarian structure, mostly relying on the use of autologous early follicles on natural or synthetic scaffolds. Based on these premises, the present study has been designed to validate the use of the ovarian bioinspired patterned electrospun fibrous scaffolds fabricated with poly(ε-caprolactone) (PCL) for multiple preantral (PA) follicle development. METHODS PA follicles isolated from lamb ovaries were cultured on PCL scaffold adopting a validated single-follicle protocol (Ctrl) or simulating a multiple-follicle condition by reproducing an artificial ovary engrafted with 5 or 10 PA (AO5PA and AO10PA). The incubations were protracted for 14 and 18 days before assessing scaffold-based microenvironment suitability to assist in vitro folliculogenesis (ivF) and oogenesis at morphological and functional level. RESULTS The ivF outcomes demonstrated that PCL-scaffolds generate an appropriate biomimetic ovarian microenvironment supporting the transition of multiple PA follicles towards early antral (EA) stage by supporting follicle growth and steroidogenic activation. PCL-multiple bioengineering ivF (AO10PA) performed in long term generated, in addition, the greatest percentage of highly specialized gametes by enhancing meiotic competence, large chromatin remodeling and parthenogenetic developmental competence. CONCLUSIONS The study showcased the proof of concept for a next-generation ART use of PCL-patterned scaffold aimed to generate transplantable artificial ovary engrafted with autologous early-stage follicles or to advance ivF technologies holding a 3D bioinspired matrix promoting a physiological long-term multiple PA follicle protocol.
Collapse
Affiliation(s)
- Chiara Di Berardino
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100, Teramo, Italy.
| | - Alessia Peserico
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100, Teramo, Italy
| | - Chiara Camerano Spelta Rapini
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100, Teramo, Italy
| | - Liliana Liverani
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Cauerstraße 6, 91058, Erlangen, Germany
- DGS SpA, Via Paolo di Dono 73, 00142, Rome, Italy
| | - Giulia Capacchietti
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100, Teramo, Italy
| | - Valentina Russo
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100, Teramo, Italy
| | - Paolo Berardinelli
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100, Teramo, Italy
| | - Irem Unalan
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Cauerstraße 6, 91058, Erlangen, Germany
| | - Andrada-Ioana Damian-Buda
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Cauerstraße 6, 91058, Erlangen, Germany
| | - Aldo R Boccaccini
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Cauerstraße 6, 91058, Erlangen, Germany
| | - Barbara Barboni
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100, Teramo, Italy
| |
Collapse
|
18
|
Wang G, Mao X, Wang W, Wang X, Li S, Wang Z. Bioprinted research models of urological malignancy. EXPLORATION (BEIJING, CHINA) 2024; 4:20230126. [PMID: 39175884 PMCID: PMC11335473 DOI: 10.1002/exp.20230126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 01/08/2024] [Indexed: 08/24/2024]
Abstract
Urological malignancy (UM) is among the leading threats to health care worldwide. Recent years have seen much investment in fundamental UM research, including mechanistic investigation, early diagnosis, immunotherapy, and nanomedicine. However, the results are not fully satisfactory. Bioprinted research models (BRMs) with programmed spatial structures and functions can serve as powerful research tools and are likely to disrupt traditional UM research paradigms. Herein, a comprehensive review of BRMs of UM is presented. It begins with a brief introduction and comparison of existing UM research models, emphasizing the advantages of BRMs, such as modeling real tissues and organs. Six kinds of mainstream bioprinting techniques used to fabricate such BRMs are summarized with examples. Thereafter, research advances in the applications of UM BRMs, such as culturing tumor spheroids and organoids, modeling cancer metastasis, mimicking the tumor microenvironment, constructing organ chips for drug screening, and isolating circulating tumor cells, are comprehensively discussed. At the end of this review, current challenges and future development directions of BRMs and UM are highlighted from the perspective of interdisciplinary science.
Collapse
Affiliation(s)
- Guanyi Wang
- Department of UrologyCancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research CenterZhongnan Hospital of Wuhan UniversityWuhanChina
- Department of Biomedical Engineering and Hubei Province Key Laboratory of Allergy and Immune Related DiseaseTaiKang Medical School (School of Basic Medical Sciences)Wuhan UniversityWuhanChina
| | - Xiongmin Mao
- Department of UrologyCancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research CenterZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Wang Wang
- Department of UrologyCancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research CenterZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Xiaolong Wang
- Lewis Katz School of MedicineTemple UniversityPhiladelphiaPennsylvaniaUSA
| | - Sheng Li
- Department of UrologyCancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research CenterZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Zijian Wang
- Department of UrologyCancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research CenterZhongnan Hospital of Wuhan UniversityWuhanChina
- Department of Biomedical Engineering and Hubei Province Key Laboratory of Allergy and Immune Related DiseaseTaiKang Medical School (School of Basic Medical Sciences)Wuhan UniversityWuhanChina
| |
Collapse
|
19
|
Akbarian M, Kianpour M, Tayebi L. Fabricating Multiphasic Angiogenic Scaffolds Using Amyloid/Roxadustat-Assisted High-Temperature Protein Printing. ACS APPLIED MATERIALS & INTERFACES 2024; 16:36983-37006. [PMID: 38953207 DOI: 10.1021/acsami.4c06207] [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: 07/03/2024]
Abstract
Repairing multiphasic defects is cumbersome. This study presents new soft and hard scaffold designs aimed at facilitating the regeneration of multiphasic defects by enhancing angiogenesis and improving cell attachment. Here, the nonimmunogenic, nontoxic, and cost-effective human serum albumin (HSA) fibril (HSA-F) was used to fabricate thermostable (up to 90 °C) and hard printable polymers. Additionally, using a 10.0 mg/mL HSA-F, an innovative hydrogel was synthesized in a mixture with 2.0% chitosan-conjugated arginine, which can gel in a cell-friendly and pH physiological environment (pH 7.4). The presence of HSA-F in both hard and soft scaffolds led to an increase in significant attachment of the scaffolds to the human periodontal ligament fibroblast (PDLF), human umbilical vein endothelial cell (HUVEC), and human osteoblast. Further studies showed that migration (up to 157%), proliferation (up to 400%), and metabolism (up to 210%) of these cells have also improved in the direction of tissue repair. By examining different in vitro and ex ovo experiments, we observed that the final multiphasic scaffold can increase blood vessel density in the process of per-vascularization as well as angiogenesis. By providing a coculture environment including PDLF and HUVEC, important cross-talk between these two cells prevails in the presence of roxadustat drug, a proangiogenic in this study. In vitro and ex ovo results demonstrated significant enhancements in the angiogenic response and cell attachment, indicating the effectiveness of the proposed design. This approach holds promise for the regeneration of complex tissue defects by providing a conducive environment for vascularization and cellular integration, thus promoting tissue healing.
Collapse
Affiliation(s)
- Mohsen Akbarian
- Marquette University School of Dentistry, Milwaukee, Wisconsin 53233, United States
| | - Maryam Kianpour
- Marquette University School of Dentistry, Milwaukee, Wisconsin 53233, United States
| | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, Wisconsin 53233, United States
| |
Collapse
|
20
|
Zhu Y, Zhang X, Chang G, Deng S, Chan HF. Bioactive Glass in Tissue Regeneration: Unveiling Recent Advances in Regenerative Strategies and Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024:e2312964. [PMID: 39014919 DOI: 10.1002/adma.202312964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 05/18/2024] [Indexed: 07/18/2024]
Abstract
Bioactive glass (BG) is a class of biocompatible, biodegradable, multifunctional inorganic glass materials, which is successfully used for orthopedic and dental applications, with several products already approved for clinical use. Apart from exhibiting osteogenic properties, BG is also known to be angiogenic and antibacterial. Recently, BG's role in immunomodulation has been gradually revealed. While the therapeutic effect of BG is mostly reported in the context of bone and skin-related regeneration, its application in regenerating other tissues/organs, such as muscle, cartilage, and gastrointestinal tissue, has also been explored recently. The strategies of applying BG have also expanded from powder or cement form to more advanced strategies such as fabrication of composite polymer-BG scaffold, 3D printing of BG-loaded scaffold, and BG-induced extracellular vesicle production. This review presents a concise overview of the recent applications of BG in regenerative medicine. Various regenerative strategies of BG will be first introduced. Next, the applications of BG in regenerating various tissues/organs, such as bone, cartilage, muscle, tendon, skin, and gastrointestinal tissue, will be discussed. Finally, summarizing clinical applications of BG for tissue regeneration will conclude, and outline future challenges and directions for the clinical translation of BG.
Collapse
Affiliation(s)
- Yanlun Zhu
- Key Laboratory for Regenerative Medicine of the Ministry of Education of China, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, P. R. China
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, P. R. China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong SAR, P. R. China
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
| | - Xuerao Zhang
- Key Laboratory for Regenerative Medicine of the Ministry of Education of China, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, P. R. China
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, P. R. China
| | - Guozhu Chang
- Key Laboratory for Regenerative Medicine of the Ministry of Education of China, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, P. R. China
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, P. R. China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong SAR, P. R. China
| | - Shuai Deng
- Key Laboratory for Regenerative Medicine of the Ministry of Education of China, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, P. R. China
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, P. R. China
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, P. R. China
| | - Hon Fai Chan
- Key Laboratory for Regenerative Medicine of the Ministry of Education of China, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, P. R. China
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, P. R. China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong SAR, P. R. China
- Hong Kong Branch of CAS Center for Excellence in Animal Evolution and Genetics, Hong Kong SAR, P. R. China
| |
Collapse
|
21
|
Nair R, Kasturi M, Mathur V, Seetharam RN, S Vasanthan K. Strategies for developing 3D printed ovarian model for restoring fertility. Clin Transl Sci 2024; 17:e13863. [PMID: 38955776 PMCID: PMC11219245 DOI: 10.1111/cts.13863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 05/16/2024] [Accepted: 05/31/2024] [Indexed: 07/04/2024] Open
Abstract
Ovaries play a crucial role in the regulation of numerous essential processes that occur within the intricate framework of female physiology. They are entrusted with the responsibility of both generating a new life and orchestrating a delicate hormonal symphony. Understanding their functioning is crucial for gaining insight into the complexities of reproduction, health, and fertility. In addition, ovaries secrete hormones that are crucial for both secondary sexual characteristics and the maintenance of overall health. A three-dimensional (3D) prosthetic ovary has the potential to restore ovarian function and preserve fertility in younger females who have undergone ovariectomies or are afflicted with ovarian malfunction. Clinical studies have not yet commenced, and the production of 3D ovarian tissue for human implantation is still in the research phase. The main challenges faced while creating a 3D ovary for in vivo implantation include sustenance of ovarian follicles, achieving vascular infiltration into the host tissue, and restoring hormone circulation. The complex ovarian microenvironment that is compartmentalized and rigid makes the biomimicking of the 3D ovary challenging in terms of biomaterial selection and bioink composition. The successful restoration of these properties in animal models has led to expectations for the development of human ovaries for implantation. This review article summarizes and evaluates the optimal 3D models of ovarian structures and their safety and efficacy concerns to provide concrete suggestions for future research.
Collapse
Affiliation(s)
- Ramya Nair
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher EducationManipalKarnatakaIndia
| | - Meghana Kasturi
- Department of Mechanical EngineeringUniversity of MichiganDearbornMichiganUSA
| | - Vidhi Mathur
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher EducationManipalKarnatakaIndia
| | - Raviraja N. Seetharam
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher EducationManipalKarnatakaIndia
| | - Kirthanashri S Vasanthan
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher EducationManipalKarnatakaIndia
| |
Collapse
|
22
|
Ngan Giang N, Le LTT, Ngoc Chien P, Trinh TTT, Thi Nga P, Zhang XR, Jin YX, Zhou SY, Han J, Nam SY, Heo CY. Assessment of inflammatory suppression and fibroblast infiltration in tissue remodelling by supercritical CO 2 acellular dermal matrix (scADM) utilizing Sprague Dawley models. Front Bioeng Biotechnol 2024; 12:1407797. [PMID: 38978716 PMCID: PMC11228881 DOI: 10.3389/fbioe.2024.1407797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 06/06/2024] [Indexed: 07/10/2024] Open
Abstract
Human skin-derived ECM aids cell functions but can trigger immune reactions; therefore it is addressed through decellularization. Acellular dermal matrices (ADMs), known for their regenerative properties, are used in tissue and organ regeneration. ADMs now play a key role in plastic and reconstructive surgery, enhancing aesthetics and reducing capsular contracture risk. Innovative decellularization with supercritical carbon dioxide preserves ECM quality for clinical use. The study investigated the cytotoxicity, biocompatibility, and anti-inflammatory properties of supercritical CO2 acellular dermal matrix (scADM) in vivo based on Sprague Dawley rat models. Initial experiments in vitro with fibroblast cells confirmed the non-toxic nature of scADM and demonstrated cell infiltration into scADMs after incubation. Subsequent tests in vitro revealed the ability of scADM to suppress inflammation induced by lipopolysaccharides (LPS) presenting by the reduction of pro-inflammatory cytokines TNF-α, IL-6, IL-1β, and MCP-1. In the in vivo model, histological assessment of implanted scADMs in 6 months revealed a decrease in inflammatory cells, confirmed further by the biomarkers of inflammation in immunofluorescence staining. Besides, an increase in fibroblast infiltration and collagen formation was observed in histological staining, which was supported by various biomarkers of fibroblasts. Moreover, the study demonstrated vascularization and macrophage polarization, depicting increased endothelial cell formation. Alteration of matrix metalloproteinases (MMPs) was analyzed by RT-PCR, indicating the reduction of MMP2, MMP3, and MMP9 levels over time. Simultaneously, an increase in collagen deposition of collagen I and collagen III was observed, verified in immunofluorescent staining, RT-PCR, and western blotting. Overall, the findings suggested that scADMs offer significant benefits in improving outcomes in implant-based procedures as well as soft tissue substitution.
Collapse
Affiliation(s)
- Nguyen Ngan Giang
- Department of Medical Device Development, College of Medicine, Seoul National University, Seoul, Republic of Korea
- Department of Plastic and Reconstructive Surgery, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Linh Thi Thuy Le
- Department of Plastic and Reconstructive Surgery, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Department of Biomedical Science, College of Medicine, Seoul National University, Seoul, Republic of Korea
- Faculty of Medical Technology, Haiphong University of Medicine and Pharmacy, Haiphong, Vietnam
| | - Pham Ngoc Chien
- Department of Plastic and Reconstructive Surgery, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Korean Institute of Nonclinical Study, H&Bio Co., Ltd., Seongnam, Republic of Korea
| | - Thuy-Tien Thi Trinh
- Department of Plastic and Reconstructive Surgery, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Korean Institute of Nonclinical Study, H&Bio Co., Ltd., Seongnam, Republic of Korea
| | - Pham Thi Nga
- Department of Plastic and Reconstructive Surgery, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Korean Institute of Nonclinical Study, H&Bio Co., Ltd., Seongnam, Republic of Korea
| | - Xin Rui Zhang
- Department of Plastic and Reconstructive Surgery, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Department of Medicine, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Yong Xun Jin
- Department of Plastic and Reconstructive Surgery, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Department of Medicine, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Shu Yi Zhou
- Department of Plastic and Reconstructive Surgery, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Department of Medicine, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | | | - Sun Young Nam
- Department of Plastic and Reconstructive Surgery, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Chan Yeong Heo
- Department of Medical Device Development, College of Medicine, Seoul National University, Seoul, Republic of Korea
- Department of Plastic and Reconstructive Surgery, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Korean Institute of Nonclinical Study, H&Bio Co., Ltd., Seongnam, Republic of Korea
- Department of Medicine, College of Medicine, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
23
|
Hernandez-Sanchez D, Comtois-Bona M, Muñoz M, Ruel M, Suuronen EJ, Alarcon EI. Manufacturing and validation of small-diameter vascular grafts: A mini review. iScience 2024; 27:109845. [PMID: 38799581 PMCID: PMC11126982 DOI: 10.1016/j.isci.2024.109845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2024] Open
Abstract
The field of small-diameter vascular grafts remains a challenge for biomaterials scientists. While decades of research have brought us much closer to developing biomimetic materials for regenerating tissues and organs, the physiological challenges involved in manufacturing small conduits that can transport blood while not inducing an immune response or promoting blood clots continue to limit progress in this area. In this short review, we present some of the most recent methods and advancements made by researchers working in the field of small-diameter vascular grafts. We also discuss some of the most critical aspects biomaterials scientists should consider when developing lab-made small-diameter vascular grafts.
Collapse
Affiliation(s)
- Deyanira Hernandez-Sanchez
- BioEngineering and Therapeutic Solutions (BEaTS) Research, Division of Cardiac Surgery, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, ON K1Y4W7, Canada
| | - Maxime Comtois-Bona
- BioEngineering and Therapeutic Solutions (BEaTS) Research, Division of Cardiac Surgery, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, ON K1Y4W7, Canada
| | - Marcelo Muñoz
- BioEngineering and Therapeutic Solutions (BEaTS) Research, Division of Cardiac Surgery, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, ON K1Y4W7, Canada
| | - Marc Ruel
- BioEngineering and Therapeutic Solutions (BEaTS) Research, Division of Cardiac Surgery, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, ON K1Y4W7, Canada
- Division of Cardiac Surgery, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, ON K1Y4W7, Canada
- Department of Cellular & Molecular Medicine, University of Ottawa, Ottawa, 451 Smyth Road, Ottawa ON K1H8M5, Canada
| | - Erik J. Suuronen
- BioEngineering and Therapeutic Solutions (BEaTS) Research, Division of Cardiac Surgery, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, ON K1Y4W7, Canada
- Department of Cellular & Molecular Medicine, University of Ottawa, Ottawa, 451 Smyth Road, Ottawa ON K1H8M5, Canada
| | - Emilio I. Alarcon
- BioEngineering and Therapeutic Solutions (BEaTS) Research, Division of Cardiac Surgery, University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, ON K1Y4W7, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H8M5, Canada
| |
Collapse
|
24
|
Sufiyan M, Kushwaha P, Ahmad M, Mandal P, Vishwakarma KK. Scaffold-Mediated Drug Delivery for Enhanced Wound Healing: A Review. AAPS PharmSciTech 2024; 25:137. [PMID: 38877197 DOI: 10.1208/s12249-024-02855-1] [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: 04/04/2024] [Accepted: 05/28/2024] [Indexed: 06/16/2024] Open
Abstract
Wound healing is a complex physiological process involving coordinated cellular and molecular events aimed at restoring tissue integrity. Acute wounds typically progress through the sequential phases of hemostasis, inflammation, proliferation, and remodeling, while chronic wounds, such as venous leg ulcers and diabetic foot ulcers, often exhibit prolonged inflammation and impaired healing. Traditional wound dressings, while widely used, have limitations such poor moisture retention and biocompatibility. To address these challenges and improve patient outcomes, scaffold-mediated delivery systems have emerged as innovative approaches. They offer advantages in creating a conducive environment for wound healing by facilitating controlled and localized drug delivery. The manuscript explores scaffold-mediated delivery systems for wound healing applications, detailing the use of natural and synthetic polymers in scaffold fabrication. Additionally, various fabrication techniques are discussed for their potential in creating scaffolds with controlled drug release kinetics. Through a synthesis of experimental findings and current literature, this manuscript elucidates the promising potential of scaffold-mediated drug delivery in improving therapeutic outcomes and advancing wound care practices.
Collapse
Affiliation(s)
- Mohd Sufiyan
- Faculty of Pharmacy, Integral University, Dasauli-Kursi Road, Lucknow, India
| | - Poonam Kushwaha
- Faculty of Pharmacy, Integral University, Dasauli-Kursi Road, Lucknow, India.
| | - Mohammad Ahmad
- Faculty of Pharmacy, Integral University, Dasauli-Kursi Road, Lucknow, India
| | - Purba Mandal
- Faculty of Pharmacy, Integral University, Dasauli-Kursi Road, Lucknow, India
| | | |
Collapse
|
25
|
Cadavid JL, Li NT, McGuigan AP. Bridging systems biology and tissue engineering: Unleashing the full potential of complex 3D in vitro tissue models of disease. BIOPHYSICS REVIEWS 2024; 5:021301. [PMID: 38617201 PMCID: PMC11008916 DOI: 10.1063/5.0179125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 03/12/2024] [Indexed: 04/16/2024]
Abstract
Rapid advances in tissue engineering have resulted in more complex and physiologically relevant 3D in vitro tissue models with applications in fundamental biology and therapeutic development. However, the complexity provided by these models is often not leveraged fully due to the reductionist methods used to analyze them. Computational and mathematical models developed in the field of systems biology can address this issue. Yet, traditional systems biology has been mostly applied to simpler in vitro models with little physiological relevance and limited cellular complexity. Therefore, integrating these two inherently interdisciplinary fields can result in new insights and move both disciplines forward. In this review, we provide a systematic overview of how systems biology has been integrated with 3D in vitro tissue models and discuss key application areas where the synergies between both fields have led to important advances with potential translational impact. We then outline key directions for future research and discuss a framework for further integration between fields.
Collapse
|
26
|
Matei MB, Marinescu CL, Matei CO, Pînzariu AS, Zăgrean L, Moisescu MG. Cost-effective optimized method to process 3D tumoral spheroids in microwell arrays for immunohistochemistry analysis. J Med Life 2024; 17:601-609. [PMID: 39296442 PMCID: PMC11407497 DOI: 10.25122/jml-2024-0267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 05/17/2024] [Indexed: 09/21/2024] Open
Abstract
This study presents an improved method for obtaining spheroids microwell arrays for histological processing and analysis, focusing on glioblastoma (U87 MG) and breast adenocarcinoma (MCF-7) tumor models. By transitioning from traditional 2D cell cultures to 3D systems, this approach overcomes the limitations of 2D cultures by more accurately replicating the tumor microenvironment. The method consists of producing homotypic and heterotypic spheroids using low-adherence agarose-coated wells, embedding these spheroids in agarose microwell arrays, and conducting immunohistochemistry (IHC) to analyze cellular and molecular profiles. Morphological analyses were performed using OrganoSeg software, and IHC staining confirmed marker expressions consistent with respective tumor types. The study details the workflow from 2D cell culture to IHC analysis, including agarose well coating, spheroid embedding, and IHC staining for markers such as EMA, p53, Ki-67, ER, PR, and HER2. Results demonstrated compact, round U87 MG spheroids and fibroblast-stabilized MCF-7 spheroids, with both types exhibiting specific marker expressions. This innovative approach significantly enhances the efficiency of producing and analyzing large volumes of spheroids, making it both quick and cost-effective. It offers a robust drug screening and cancer research platform, maintaining spheroid traceability even in bulk workflow conditions. Furthermore, this methodology supports advances in personalized medicine by providing a more physiologically relevant model than 2D cultures, which is crucial for investigating tumor behavior and therapeutic responses through IHC.
Collapse
Affiliation(s)
- Mircea Bogdan Matei
- Physiology and Neuroscience Department, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
- Biophysics and Cellular Biotechnology Department, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Carmen Letitia Marinescu
- Biophysics and Cellular Biotechnology Department, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
- Pathology Department, Sante Medical Clinic, Bucharest, Romania
| | - Christien Oktaviani Matei
- Biophysics and Cellular Biotechnology Department, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
- Excellence Center for Research in Biophysics and Cellular Biotechnology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | | | - Leon Zăgrean
- Physiology and Neuroscience Department, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Mihaela Georgeta Moisescu
- Biophysics and Cellular Biotechnology Department, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
- Excellence Center for Research in Biophysics and Cellular Biotechnology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| |
Collapse
|
27
|
Kesharwani P, Alexander A, Shukla R, Jain S, Bisht A, Kumari K, Verma K, Sharma S. Tissue regeneration properties of hydrogels derived from biological macromolecules: A review. Int J Biol Macromol 2024; 271:132280. [PMID: 38744364 DOI: 10.1016/j.ijbiomac.2024.132280] [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: 01/13/2024] [Revised: 05/02/2024] [Accepted: 05/08/2024] [Indexed: 05/16/2024]
Abstract
The successful tissue engineering depends on the development of biologically active scaffolds that possess optimal characteristics to effectively support cellular functions, maintain structural integrity and aid in tissue regeneration. Hydrogels have emerged as promising candidates in tissue regeneration due to their resemblance to the natural extracellular matrix and their ability to support cell survival and proliferation. The integration of hydrogel scaffold into the polymer has a variable impact on the pseudo extracellular environment, fostering cell growth/repair. The modification in size, shape, surface morphology and porosity of hydrogel scaffolds has consequently paved the way for addressing diverse challenges in the tissue engineering process such as tissue architecture, vascularization and simultaneous seeding of multiple cells. The present review provides a comprehensive update on hydrogel production using natural and synthetic biomaterials and their underlying mechanisms. Furthermore, it delves into the application of hydrogel scaffolds in tissue engineering for cardiac tissues, cartilage tissue, adipose tissue, nerve tissue and bone tissue. Besides, the present article also highlights various clinical studies, patents, and the limitations associated with hydrogel-based scaffolds in recent times.
Collapse
Affiliation(s)
- Payal Kesharwani
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, Rajasthan, India; Institute of Pharmacy, Ram-Eesh Institute of Vocational and Technical Education Greater Noida, India
| | - Amit Alexander
- Department of Pharmaceuticals, National Institute of Pharmaceutical Education and Research, Guwahati, Assam, India
| | - Rahul Shukla
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Raebareli, Lucknow, Uttar Pradesh, India
| | - Smita Jain
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, Rajasthan, India
| | - Akansha Bisht
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, Rajasthan, India
| | - Kajal Kumari
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, Rajasthan, India
| | - Kanika Verma
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, Rajasthan, India
| | - Swapnil Sharma
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, Rajasthan, India.
| |
Collapse
|
28
|
Cui L, Pi J, Qin B, Cui T, Liu Z, Lei L, Wu S. Advanced application of carbohydrate-based micro/nanoparticles for rheumatoid arthritis. Int J Biol Macromol 2024; 269:131809. [PMID: 38677672 DOI: 10.1016/j.ijbiomac.2024.131809] [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: 12/31/2023] [Revised: 04/03/2024] [Accepted: 04/05/2024] [Indexed: 04/29/2024]
Abstract
Rheumatoid arthritis (RA) is a kind of synovitis and progressive joint destruction disease. Dysregulated immune cell activation, inflammatory cytokine overproduction, and subsequent reactive oxidative species (ROS) production contribute to the RA process. Carbohydrates, including cellulose, chitosan, alginate and dextran, are among the most abundant and important biomolecules in nature and are widely used in biomedicine. Carbohydrate-based micro/nanoparticles(M/NPs) as functional excipients have the ability to improve the bioavailability, solubility and stability of numerous drugs used in RA therapy. For on-demand therapy, smart reactive M/NPs have been developed to respond to a variety of chemical and physical stimuli, including light, temperature, enzymes, pH and ROS, alternating their physical and macroscopic properties, resulting in innovative new drug delivery systems. In particular, advanced products with targeted dextran or hyaluronic acid are exploiting multiple beneficial properties at the same time. In addition to those that respond, there are promising new derivatives in development with microenvironment and chronotherapy effects. In this review, we provide an overview of these recent developments and an outlook on how this class of agents will further shape the landscape of drug delivery for RA treatment.
Collapse
Affiliation(s)
- Linxian Cui
- Geriatric Diseases Institute of Chengdu/Cancer Prevention and Treatment Institute of Chengdu, Department of Cardiology, Chengdu Fifth People's Hospital (The Second Clinical Medical College, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, Sichuan 611130, PR China
| | - Jinkui Pi
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Boquan Qin
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Ting Cui
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Zhenfei Liu
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Lei Lei
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, PR China.
| | - Shizhou Wu
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China.
| |
Collapse
|
29
|
Razavi ZS, Soltani M, Mahmoudvand G, Farokhi S, Karimi-Rouzbahani A, Farasati-Far B, Tahmasebi-Ghorabi S, Pazoki-Toroudi H, Afkhami H. Advancements in tissue engineering for cardiovascular health: a biomedical engineering perspective. Front Bioeng Biotechnol 2024; 12:1385124. [PMID: 38882638 PMCID: PMC11176440 DOI: 10.3389/fbioe.2024.1385124] [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: 02/17/2024] [Accepted: 05/13/2024] [Indexed: 06/18/2024] Open
Abstract
Myocardial infarction (MI) stands as a prominent contributor to global cardiovascular disease (CVD) mortality rates. Acute MI (AMI) can result in the loss of a large number of cardiomyocytes (CMs), which the adult heart struggles to replenish due to its limited regenerative capacity. Consequently, this deficit in CMs often precipitates severe complications such as heart failure (HF), with whole heart transplantation remaining the sole definitive treatment option, albeit constrained by inherent limitations. In response to these challenges, the integration of bio-functional materials within cardiac tissue engineering has emerged as a groundbreaking approach with significant potential for cardiac tissue replacement. Bioengineering strategies entail fortifying or substituting biological tissues through the orchestrated interplay of cells, engineering methodologies, and innovative materials. Biomaterial scaffolds, crucial in this paradigm, provide the essential microenvironment conducive to the assembly of functional cardiac tissue by encapsulating contracting cells. Indeed, the field of cardiac tissue engineering has witnessed remarkable strides, largely owing to the application of biomaterial scaffolds. However, inherent complexities persist, necessitating further exploration and innovation. This review delves into the pivotal role of biomaterial scaffolds in cardiac tissue engineering, shedding light on their utilization, challenges encountered, and promising avenues for future advancement. By critically examining the current landscape, we aim to catalyze progress toward more effective solutions for cardiac tissue regeneration and ultimately, improved outcomes for patients grappling with cardiovascular ailments.
Collapse
Affiliation(s)
- Zahra-Sadat Razavi
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Madjid Soltani
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
- Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, ON, Canada
- Centre for Sustainable Business, International Business University, Toronto, ON, Canada
| | - Golnaz Mahmoudvand
- Student Research Committee, USERN Office, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Simin Farokhi
- Student Research Committee, USERN Office, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Arian Karimi-Rouzbahani
- Student Research Committee, USERN Office, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Bahareh Farasati-Far
- Department of Chemistry, Iran University of Science and Technology, Tehran, Iran
| | - Samaneh Tahmasebi-Ghorabi
- Master of Health Education, Research Expert, Clinical Research Development Unit, Emam Khomeini Hospital, Ilam University of Medical Sciences, Ilam, Iran
| | | | - Hamed Afkhami
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| |
Collapse
|
30
|
Moghaddam A, Bahrami M, Mirzadeh M, Khatami M, Simorgh S, Chimehrad M, Kruppke B, Bagher Z, Mehrabani D, Khonakdar HA. Recent trends in bone tissue engineering: a review of materials, methods, and structures. Biomed Mater 2024; 19:042007. [PMID: 38636500 DOI: 10.1088/1748-605x/ad407d] [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: 09/23/2023] [Accepted: 04/18/2024] [Indexed: 04/20/2024]
Abstract
Bone tissue engineering (BTE) provides the treatment possibility for segmental long bone defects that are currently an orthopedic dilemma. This review explains different strategies, from biological, material, and preparation points of view, such as using different stem cells, ceramics, and metals, and their corresponding properties for BTE applications. In addition, factors such as porosity, surface chemistry, hydrophilicity and degradation behavior that affect scaffold success are introduced. Besides, the most widely used production methods that result in porous materials are discussed. Gene delivery and secretome-based therapies are also introduced as a new generation of therapies. This review outlines the positive results and important limitations remaining in the clinical application of novel BTE materials and methods for segmental defects.
Collapse
Affiliation(s)
| | - Mehran Bahrami
- Department of Mechanical Engineering and Mechanics, Lehigh University, 27 Memorial Dr W, Bethlehem, PA 18015, United States of America
| | | | - Mehrdad Khatami
- Iran Polymer and Petrochemical Institute (IPPI), Tehran 14965-115, Iran
| | - Sara Simorgh
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Chimehrad
- Department of Mechanical & Aerospace Engineering, College of Engineering & Computer Science, University of Central Florida, Orlando, FL, United States of America
| | - Benjamin Kruppke
- Max Bergmann Center of Biomaterials and Institute of Materials Science, Technische Universität Dresden, 01069 Dresden, Germany
| | - Zohreh Bagher
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Davood Mehrabani
- Burn and Wound Healing Research Center, Shiraz University of Medical Sciences, Shiraz, Fars 71348-14336, Iran
- Stem Cell Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Fars 71345-1744, Iran
| | - Hossein Ali Khonakdar
- Iran Polymer and Petrochemical Institute (IPPI), Tehran 14965-115, Iran
- Max Bergmann Center of Biomaterials and Institute of Materials Science, Technische Universität Dresden, 01069 Dresden, Germany
| |
Collapse
|
31
|
Kumar P, Shamim, Muztaba M, Ali T, Bala J, Sidhu HS, Bhatia A. Fused Deposition Modeling 3D-Printed Scaffolds for Bone Tissue Engineering Applications: A Review. Ann Biomed Eng 2024; 52:1184-1194. [PMID: 38418691 DOI: 10.1007/s10439-024-03479-z] [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: 01/22/2024] [Accepted: 02/16/2024] [Indexed: 03/02/2024]
Abstract
The emergence of bone tissue engineering as a trend in regenerative medicine is forcing scientists to create highly functional materials and scaffold construction techniques. Bone tissue engineering uses 3D bio-printed scaffolds that allow and stimulate the attachment and proliferation of osteoinductive cells on their surfaces. Bone grafting is necessary to expedite the patient's condition because the natural healing process of bones is slow. Fused deposition modeling (FDM) is therefore suggested as a technique for the production process due to its simplicity, ability to create intricate components and movable forms, and low running costs. 3D-printed scaffolds can repair bone defects in vivo and in vitro. For 3D printing, various materials including metals, polymers, and ceramics are often employed but polymeric biofilaments are promising candidates for replacing non-biodegradable materials due to their adaptability and environment friendliness. This review paper majorly focuses on the fused deposition modeling approach for the fabrication of 3D scaffolds. In addition, it also provides information on biofilaments used in FDM 3D printing, applications, and commercial aspects of scaffolds in bone tissue engineering.
Collapse
Affiliation(s)
- Pawan Kumar
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, 151001, India.
| | - Shamim
- IIMT College of Medical Sciences, IIMT University, Ganga Nagar, Meerut, Uttar Pradesh, 250001, India
| | - Mohammad Muztaba
- Department of Pharmacology, Praduman Singh Sikshan Prasikshan Sansthan Pharmacy College, Phutahiya Sansarpur, Basti, Uttar Pradesh, 272001, India
| | - Tarmeen Ali
- Department of Pharmacy, Swami Vivekanand Subharti University, Subhartipuram, Meerut, Uttar Pradesh, 250005, India
| | - Jyoti Bala
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, 151001, India
| | - Haramritpal Singh Sidhu
- Department of Mechanical Engineering, Giani Zail Singh Campus College of Engineering & Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, 151001, India
| | - Amit Bhatia
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, 151001, India
| |
Collapse
|
32
|
Han Y, Wu Y, Wang F, Li G, Wang J, Wu X, Deng A, Ren X, Wang X, Gao J, Shi Z, Bai L, Su J. Heterogeneous DNA hydrogel loaded with Apt02 modified tetrahedral framework nucleic acid accelerated critical-size bone defect repair. Bioact Mater 2024; 35:1-16. [PMID: 38298451 PMCID: PMC10828543 DOI: 10.1016/j.bioactmat.2024.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/10/2024] [Accepted: 01/10/2024] [Indexed: 02/02/2024] Open
Abstract
Segmental bone defects, stemming from trauma, infection, and tumors, pose formidable clinical challenges. Traditional bone repair materials, such as autologous and allogeneic bone grafts, grapple with limitations including source scarcity and immune rejection risks. The advent of nucleic acid nanotechnology, particularly the use of DNA hydrogels in tissue engineering, presents a promising solution, attributed to their biocompatibility, biodegradability, and programmability. However, these hydrogels, typically hindered by high gelation temperatures (∼46 °C) and high construction costs, limit cell encapsulation and broader application. Our research introduces a novel polymer-modified DNA hydrogel, developed using nucleic acid nanotechnology, which gels at a more biocompatible temperature of 37 °C and is cost-effective. This hydrogel then incorporates tetrahedral Framework Nucleic Acid (tFNA) to enhance osteogenic mineralization. Furthermore, considering the modifiability of tFNA, we modified its chains with Aptamer02 (Apt02), an aptamer known to foster angiogenesis. This dual approach significantly accelerates osteogenic differentiation in bone marrow stromal cells (BMSCs) and angiogenesis in human umbilical vein endothelial cells (HUVECs), with cell sequencing confirming their targeting efficacy, respectively. In vivo experiments in rats with critical-size cranial bone defects demonstrate their effectiveness in enhancing new bone formation. This innovation not only offers a viable solution for repairing segmental bone defects but also opens avenues for future advancements in bone organoids construction, marking a significant advancement in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Yafei Han
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- School of Medicine, Shanghai University, Shanghai, 200444, China
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China
| | - Yan Wu
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Fuxiao Wang
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- School of Medicine, Shanghai University, Shanghai, 200444, China
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China
| | - Guangfeng Li
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- School of Medicine, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai, 200444, China
| | - Jian Wang
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- School of Medicine, Shanghai University, Shanghai, 200444, China
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China
| | - Xiang Wu
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Anfu Deng
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Xiaoxiang Ren
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Xiuhui Wang
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Jie Gao
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Zhongmin Shi
- National Center for Orthopaedics, Shanghai Sixth People's Hospital, Shanghai, 200233, China
| | - Long Bai
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- Wenzhou Institute of Shanghai University, Wenzhou, 325000, China
| | - Jiacan Su
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| |
Collapse
|
33
|
Chen W, Wu P, Jin C, Chen Y, Li C, Qian H. Advances in the application of extracellular vesicles derived from three-dimensional culture of stem cells. J Nanobiotechnology 2024; 22:215. [PMID: 38693585 PMCID: PMC11064407 DOI: 10.1186/s12951-024-02455-y] [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: 01/05/2024] [Accepted: 04/02/2024] [Indexed: 05/03/2024] Open
Abstract
Stem cells (SCs) have been used therapeutically for decades, yet their applications are limited by factors such as the risk of immune rejection and potential tumorigenicity. Extracellular vesicles (EVs), a key paracrine component of stem cell potency, overcome the drawbacks of stem cell applications as a cell-free therapeutic agent and play an important role in treating various diseases. However, EVs derived from two-dimensional (2D) planar culture of SCs have low yield and face challenges in large-scale production, which hinders the clinical translation of EVs. Three-dimensional (3D) culture, given its ability to more realistically simulate the in vivo environment, can not only expand SCs in large quantities, but also improve the yield and activity of EVs, changing the content of EVs and improving their therapeutic effects. In this review, we briefly describe the advantages of EVs and EV-related clinical applications, provide an overview of 3D cell culture, and finally focus on specific applications and future perspectives of EVs derived from 3D culture of different SCs.
Collapse
Affiliation(s)
- Wenya Chen
- Department of Orthopaedics, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, 215300, Jiangsu, China
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
| | - Peipei Wu
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Can Jin
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
| | - Yinjie Chen
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
| | - Chong Li
- Department of Orthopaedics, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, 215300, Jiangsu, China.
| | - Hui Qian
- Department of Orthopaedics, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, 215300, Jiangsu, China.
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China.
| |
Collapse
|
34
|
Limpiwatana S, Nagaviroj N. Intaglio Surface Adaptation of Removable Partial Denture Framework Fabricated by Various Data Acquisition Techniques and Fabrication Approaches. Eur J Dent 2024; 18:493-500. [PMID: 37729933 PMCID: PMC11132788 DOI: 10.1055/s-0043-1772245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2023] Open
Abstract
OBJECTIVES The aim of this study was to compare intaglio surface adaptation of the removable partial denture framework among various data acquisition techniques and fabrication approaches using three-dimensional comparison by metrology software. MATERIALS AND METHODS The partial edentulous typodont model with five digital superimposition landmarks was duplicated and scanned for the digital reference model. Three approaches were the conventional lost-wax (group I; LWT, n = 5), intraoral digital impressions combined with PolyJet printing and lost-wax (group II; IP-LWT, n = 5), and extraoral digital impressions combined with PolyJet printing and lost-wax (group III; EP-LWT, n = 5). Each framework was scanned and superimposed with the reference model. The misfits at 53 locations were measured. STATISTICAL ANALYSIS Data were statistically analyzed by one-way analysis of variance, followed by Tukey's honestly significant difference for pairwise comparisons (p < 0.05). RESULTS Significant differences were found between three approaches at the reciprocal arm, terminal part of the retentive arm, rest, and major connector (p < 0.05). In the LWT group, the reciprocal arm and palatal vault region of major connector had the lowest misfits, but the highest misfit was found in the midline region (p < 0.001). In the IP-LWT group revealed the most excessive contact at the terminal part of the retentive arm (-0.111 ± 0.038 mm, p = 0.031), with the highest misfit at the rest area (p < 0.001). CONCLUSION A difference in adaptation was found in several removable partial denture framework components among three approaches. The LWT group had a better adaptation than other groups. Nevertheless, a clinically acceptable adaptation was seen in all three approaches.
Collapse
Affiliation(s)
- Seehachart Limpiwatana
- Residency Training in Prosthodontics, Department of Prosthodontics, Faculty of Dentistry, Mahidol University, Bangkok, Thailand
| | - Noppavan Nagaviroj
- Department of Prosthodontics, Faculty of Dentistry, Mahidol University, Bangkok, Thailand
| |
Collapse
|
35
|
Zhou H, Zhang Z, Mu Y, Yao H, Zhang Y, Wang DA. Harnessing Nanomedicine for Cartilage Repair: Design Considerations and Recent Advances in Biomaterials. ACS NANO 2024; 18:10667-10687. [PMID: 38592060 DOI: 10.1021/acsnano.4c00780] [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: 04/10/2024]
Abstract
Cartilage injuries are escalating worldwide, particularly in aging society. Given its limited self-healing ability, the repair and regeneration of damaged articular cartilage remain formidable challenges. To address this issue, nanomaterials are leveraged to achieve desirable repair outcomes by enhancing mechanical properties, optimizing drug loading and bioavailability, enabling site-specific and targeted delivery, and orchestrating cell activities at the nanoscale. This review presents a comprehensive survey of recent research in nanomedicine for cartilage repair, with a primary focus on biomaterial design considerations and recent advances. The review commences with an introductory overview of the intricate cartilage microenvironment and further delves into key biomaterial design parameters crucial for treating cartilage damage, including microstructure, surface charge, and active targeting. The focal point of this review lies in recent advances in nano drug delivery systems and nanotechnology-enabled 3D matrices for cartilage repair. We discuss the compositions and properties of these nanomaterials and elucidate how these materials impact the regeneration of damaged cartilage. This review underscores the pivotal role of nanotechnology in improving the efficacy of biomaterials utilized for the treatment of cartilage damage.
Collapse
Affiliation(s)
- Huiqun Zhou
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China
| | - Zhen Zhang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China
| | - Yulei Mu
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China
| | - Hang Yao
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou 225009, China
| | - Yi Zhang
- School of Integrated Circuit Science and Engineering, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Dong-An Wang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China
- Center for Neuromusculoskeletal Restorative Medicine, InnoHK, HKSTP, Sha Tin, Hong Kong SAR 999077, China
| |
Collapse
|
36
|
Iravani S, Nazarzadeh Zare E, Makvandi P. Multifunctional MXene-Based Platforms for Soft and Bone Tissue Regeneration and Engineering. ACS Biomater Sci Eng 2024; 10:1892-1909. [PMID: 38466909 DOI: 10.1021/acsbiomaterials.3c01770] [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: 03/13/2024]
Abstract
MXenes and their composites hold great promise in the field of soft and bone tissue regeneration and engineering (TRE). However, there are challenges that need to be overcome, such as ensuring biocompatibility and controlling the morphologies of MXene-based scaffolds. The future prospects of MXenes in TRE include enhancing biocompatibility through surface modifications, developing multifunctional constructs, and conducting in vivo studies for clinical translation. The purpose of this perspective about MXenes and their composites in soft and bone TRE is to critically evaluate their potential applications and contributions in this field. This perspective aims to provide a comprehensive analysis of the challenges, advantages, limitations, and future prospects associated with the use of MXenes and their composites for soft and bone TRE. By examining the existing literature and research, the review seeks to consolidate the current knowledge and highlight the key findings and advancements in MXene-based TRE. It aims to contribute to the understanding of MXenes' role in promoting soft and bone TRE, addressing the challenges faced in terms of biocompatibility, morphology control, and tissue interactions.
Collapse
Affiliation(s)
- Siavash Iravani
- Independent Researcher, W Nazar ST, Boostan Avenue, Isfahan 81756-33551, Iran
| | - Ehsan Nazarzadeh Zare
- School of Chemistry, Damghan University, Damghan 36716-45667, Iran
- Centre of Research Impact and Outreach, Chitkara University, Rajpura 140417, Punjab, India
| | - Pooyan Makvandi
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou 324000, Zhejiang, China
- Chitkara Centre for Research and Development, Chitkara University, Kalujhanda 174103, Himachal Pradesh, India
- Department of Biomaterials, Saveetha Dental College and Hospitals, SIMATS, Saveetha University, Chennai 600077, India
| |
Collapse
|
37
|
Das S, Jegadeesan JT, Basu B. Gelatin Methacryloyl (GelMA)-Based Biomaterial Inks: Process Science for 3D/4D Printing and Current Status. Biomacromolecules 2024; 25:2156-2221. [PMID: 38507816 DOI: 10.1021/acs.biomac.3c01271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
Tissue engineering for injured tissue replacement and regeneration has been a subject of investigation over the last 30 years, and there has been considerable interest in using additive manufacturing to achieve these goals. Despite such efforts, many key questions remain unanswered, particularly in the area of biomaterial selection for these applications as well as quantitative understanding of the process science. The strategic utilization of biological macromolecules provides a versatile approach to meet diverse requirements in 3D printing, such as printability, buildability, and biocompatibility. These molecules play a pivotal role in both physical and chemical cross-linking processes throughout the biofabrication, contributing significantly to the overall success of the 3D printing process. Among the several bioprintable materials, gelatin methacryloyl (GelMA) has been widely utilized for diverse tissue engineering applications, with some degree of success. In this context, this review will discuss the key bioengineering approaches to identify the gelation and cross-linking strategies that are appropriate to control the rheology, printability, and buildability of biomaterial inks. This review will focus on the GelMA as the structural (scaffold) biomaterial for different tissues and as a potential carrier vehicle for the transport of living cells as well as their maintenance and viability in the physiological system. Recognizing the importance of printability toward shape fidelity and biophysical properties, a major focus in this review has been to discuss the qualitative and quantitative impact of the key factors, including microrheological, viscoelastic, gelation, shear thinning properties of biomaterial inks, and printing parameters, in particular, reference to 3D extrusion printing of GelMA-based biomaterial inks. Specifically, we emphasize the different possibilities to regulate mechanical, swelling, biodegradation, and cellular functionalities of GelMA-based bio(material) inks, by hybridization techniques, including different synthetic and natural biopolymers, inorganic nanofillers, and microcarriers. At the close, the potential possibility of the integration of experimental data sets and artificial intelligence/machine learning approaches is emphasized to predict the printability, shape fidelity, or biophysical properties of GelMA bio(material) inks for clinically relevant tissues.
Collapse
Affiliation(s)
- Soumitra Das
- Materials Research Centre, Indian Institute of Science, Bangalore, India 560012
| | | | - Bikramjit Basu
- Materials Research Centre, Indian Institute of Science, Bangalore, India 560012
| |
Collapse
|
38
|
Fromager B, Cambedouzou J, Marhuenda E, Iskratsch T, Pinault M, Bakalara N, Cornu D. Tunable electrospun scaffolds of polyacrylonitrile loaded with carbon nanotubes: from synthesis to biological applications. Chembiochem 2024; 25:e202300768. [PMID: 38353030 DOI: 10.1002/cbic.202300768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 02/03/2024] [Indexed: 03/13/2024]
Abstract
Growing cells in a biomimetic environment is critical for tissue engineering as well as for studying the cell biology underlying disease mechanisms. To this aim a range of 3D matrices have been developed, from hydrogels to decellularized matrices. They need to mimic the extracellular matrix to ensure the optimal growth and function of cells. Electrospinning has gained in popularity due to its capacity to individually tune chemistry and mechanical properties and as such influence cell attachment, differentiation or maturation. Polyacrylonitrile (PAN) derived electrospun fibres scaffolds have shown exciting potential due to reports of mechanical tunability and biocompatibility. Building on previous work we fabricate here a range of PAN fibre scaffolds with different concentrations of carbon nanotubes. We characterize them in-depth in respect to their structure, surface chemistry and mechanical properties, using scanning electron microscopy, image processing, ultramicrotomic transmission electron microscopy, x-ray nanotomography, infrared spectroscopy, atomic force microscopy and nanoindentation. Together the data demonstrate this approach to enable finetuning the mechanical properties, while keeping the structure and chemistry unaltered and hence offering ideal properties for comparative studies of the cellular mechanobiology. Finally, we confirm the biocompatibility of the scaffolds using primary rat cardiomyocytes, vascular smooth muscle (A7r5) and myoblast (C2C12) cell lines.
Collapse
Affiliation(s)
- Bénédicte Fromager
- IEM, Univ Montpellier, CNRS, ENSCM, cc047 Pl. E. Bataillon, 34095, Montpellier, France
| | - Julien Cambedouzou
- IEM, Univ Montpellier, CNRS, ENSCM, cc047 Pl. E. Bataillon, 34095, Montpellier, France
| | - Emilie Marhuenda
- School of Engineering and Materials Science, Queen Mary University Of London, 327 Mile End Rd, Bethnal Green, London, E1 4NS, Royaume-Uni
| | - Thomas Iskratsch
- School of Engineering and Materials Science, Queen Mary University Of London, 327 Mile End Rd, Bethnal Green, London, E1 4NS, Royaume-Uni
| | - Mathieu Pinault
- Univ Paris Saclay, CEA, CNRS, NIMBE,LEDNA, F-91191, Gif Sur Yvette, France
| | - Norbert Bakalara
- CNRS, ENSTBB-Bordeaux INP, Université de Bordeaux, 146 rue Léo Saignat, 33076, Bordeaux, France
| | - David Cornu
- IEM, Univ Montpellier, CNRS, ENSCM, cc047 Pl. E. Bataillon, 34095, Montpellier, France
| |
Collapse
|
39
|
Javkhlan Z, Hsu SH, Chen RS, Chen MH. Interactions of neural-like cells with 3D-printed polycaprolactone with different inner diameters for neural regeneration. J Dent Sci 2024; 19:1096-1104. [PMID: 38618126 PMCID: PMC11010800 DOI: 10.1016/j.jds.2023.12.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 12/20/2023] [Indexed: 04/16/2024] Open
Abstract
Background/purpose Peripheral neural regeneration is an interesting and challenging field. The aim of this study was to investigate the interactions of neural-like PC12 cells and Poly-D-Lysine (PDL)-coated 3D-printed polycaprolactone (PCL) scaffolds with different inner diameters of half tubular array (HTA) (0, 200, 300, and 400 μm), respectively. Materials and methods This study used the fused deposition modeling (FDM) technique with 3D-printing to fabricate the thermoplastic polymer. Scaffold properties were measured by mechanical testing, and coating quality was observed under a scanning electron microscope (SEM). PC12 cell biocompatibility was examined by an MTT assay. Cell differentiation was evaluated by immunofluorescence staining. Results The cell viability of PC12 cells on PDL-coated PCL scaffolds with a 200-μm inner diameter of HTA was shown with significant differences (∗P < 0.05, ∗∗P < 0.01, ∗∗∗P < 0.001) than other PCL groups at all experimental dates. The SEM observation showed that PDL-coated PCL scaffolds with 200-μm inner diameters of HTA promoted cell adhesion. An immunofluorescence staining of PC12 cells on the PDL-coated PCL scaffold with a 200-μm inner diameter of the HTA group showed that it stimulated PC12 cells for neurite formation much better than the other groups.A PDL-coated PCL scaffold with a 200-μm inner diameter of HTA can promote the growth and differentiation of PC12 cells better than other groups. It indicated that PDL-coated PCL scaffolds with a 200-μm inner diameter HTA can be used for further neural regeneration application.
Collapse
Affiliation(s)
- Zolzaya Javkhlan
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan
| | - Sheng-Hao Hsu
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan
| | - Rung-Shu Chen
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan
| | - Min-Huey Chen
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan
- Department of Dentistry, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
40
|
De Vitis E, Stanzione A, Romano A, Quattrini A, Gigli G, Moroni L, Gervaso F, Polini A. The Evolution of Technology-Driven In Vitro Models for Neurodegenerative Diseases. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304989. [PMID: 38366798 DOI: 10.1002/advs.202304989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 01/15/2024] [Indexed: 02/18/2024]
Abstract
The alteration in the neural circuits of both central and peripheral nervous systems is closely related to the onset of neurodegenerative disorders (NDDs). Despite significant research efforts, the knowledge regarding NDD pathological processes, and the development of efficacious drugs are still limited due to the inability to access and reproduce the components of the nervous system and its intricate microenvironment. 2D culture systems are too simplistic to accurately represent the more complex and dynamic situation of cells in vivo and have therefore been surpassed by 3D systems. However, both models suffer from various limitations that can be overcome by employing two innovative technologies: organ-on-chip and 3D printing. In this review, an overview of the advantages and shortcomings of both microfluidic platforms and extracellular matrix-like biomaterials will be given. Then, the combination of microfluidics and hydrogels as a new synergistic approach to study neural disorders by analyzing the latest advances in 3D brain-on-chip for neurodegenerative research will be explored.
Collapse
Affiliation(s)
- Eleonora De Vitis
- CNR NANOTEC-Institute of Nanotechnology, Campus Ecotekn, via Monteroni, Lecce, 73100, Italy
| | - Antonella Stanzione
- CNR NANOTEC-Institute of Nanotechnology, Campus Ecotekn, via Monteroni, Lecce, 73100, Italy
| | - Alessandro Romano
- IRCCS San Raffaele Scientific Institute, Division of Neuroscience, Institute of Experimental Neurology, Milan, 20132, Italy
| | - Angelo Quattrini
- IRCCS San Raffaele Scientific Institute, Division of Neuroscience, Institute of Experimental Neurology, Milan, 20132, Italy
| | - Giuseppe Gigli
- CNR NANOTEC-Institute of Nanotechnology, Campus Ecotekn, via Monteroni, Lecce, 73100, Italy
- Dipartimento di Medicina Sperimentale, Università Del Salento, Campus Ecotekne, via Monteroni, Lecce, 73100, Italy
| | - Lorenzo Moroni
- CNR NANOTEC-Institute of Nanotechnology, Campus Ecotekn, via Monteroni, Lecce, 73100, Italy
- Complex Tissue Regeneration, Maastricht University, Universiteitssingel 40, Maastricht, 6229 ER, Netherlands
| | - Francesca Gervaso
- CNR NANOTEC-Institute of Nanotechnology, Campus Ecotekn, via Monteroni, Lecce, 73100, Italy
| | - Alessandro Polini
- CNR NANOTEC-Institute of Nanotechnology, Campus Ecotekn, via Monteroni, Lecce, 73100, Italy
| |
Collapse
|
41
|
Kim MJ, Park H, Jung R, Won C, Ohk S, Kim H, Roh N, Yi K. High-resolution 3-D scanning electron microscopy (SEM) images of DOT TM polynucleotides (PN): Unique scaffold characteristics and potential applications in biomedicine. Skin Res Technol 2024; 30:e13667. [PMID: 38558437 PMCID: PMC10982675 DOI: 10.1111/srt.13667] [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: 03/06/2024] [Accepted: 03/11/2024] [Indexed: 04/04/2024]
Abstract
INTRODUCTION Polynucleotides (PN) are becoming more prominent in aesthetic medicine. However, the structural characteristics of PN have not been published and PN from different companies may have different structural characteristics. This study aimed to elucidate the structural attributes of DOT™ PN and distinguish differences with polydeoxyribonucleotides (PDRN) using high-resolution scanning electron microscopy (SEM) imaging. MATERIALS AND METHODS DOT™ PN was examined using a Quanta 3-D field emission gun (FEG) Scanning Electron Microscope (SEM). Sample preparation involved cryogenic cooling, cleavage, etching, and metal coating to facilitate high-resolution imaging. Cryo-FIB/SEM techniques were employed for in-depth structural analysis. RESULTS PDRN exhibited an amorphous structure without distinct features. In contrast, DOT™ PN displayed well-defined polyhedral shapes with smooth, uniformly thick walls. These cells were empty, with diameters ranging from 3 to 8 micrometers, forming a seamless tessellation pattern. DISCUSSION DOT™ PN's distinct geometric tessellation design conforms to the principles of biotensegrity, providing both structural reinforcement and integrity. The presence of delicate partitions and vacant compartments hints at possible uses in the field of pharmaceutical delivery systems. Within the realms of beauty enhancement and regenerative medicine, DOT™ PN's capacity to bolster cell growth and tissue mending could potentially transform approaches to rejuvenation treatments. Its adaptability becomes apparent when considering its contributions to drug administration and surgical procedures. CONCLUSION This study unveils the intricate structural scaffold features of DOT™ PN for the first time, setting it apart from PDRN and inspiring innovation in biomedicine and materials science. DOT™ PN's unique attributes open doors to potential applications across healthcare and beyond.
Collapse
Affiliation(s)
| | | | - Rae‐Jun Jung
- Pharmaresearch Co., Ltd. Integrated R&D CenterSungnamSouth Korea
| | - Chee‐Youb Won
- Pharmaresearch Co., Ltd. Integrated R&D CenterSungnamSouth Korea
| | - Seul‐Ong Ohk
- Pharmaresearch Co., Ltd. Integrated R&D CenterSungnamSouth Korea
| | - Hong‐Taek Kim
- Pharmaresearch Co., Ltd. Integrated R&D CenterSungnamSouth Korea
| | - Nark‐Kyung Roh
- Leaders Aesthetic Laser and Cosmetic Surgery CenterSeoulSouth Korea
| | - Kyu‐Ho Yi
- Maylin Clinic (Apgujeong)SeoulSouth Korea
- Division in Anatomy and Developmental BiologyDepartment of Oral BiologyHuman Identification Research InstituteBK21 FOUR ProjectYonsei University College of DentistrySeoulSouth Korea
| |
Collapse
|
42
|
Patil R, Alimperti S. Graphene in 3D Bioprinting. J Funct Biomater 2024; 15:82. [PMID: 38667539 PMCID: PMC11051043 DOI: 10.3390/jfb15040082] [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: 02/22/2024] [Revised: 03/14/2024] [Accepted: 03/19/2024] [Indexed: 04/28/2024] Open
Abstract
Three-dimensional (3D) bioprinting is a fast prototyping fabrication approach that allows the development of new implants for tissue restoration. Although various materials have been utilized for this process, they lack mechanical, electrical, chemical, and biological properties. To overcome those limitations, graphene-based materials demonstrate unique mechanical and electrical properties, morphology, and impermeability, making them excellent candidates for 3D bioprinting. This review summarizes the latest developments in graphene-based materials in 3D printing and their application in tissue engineering and regenerative medicine. Over the years, different 3D printing approaches have utilized graphene-based materials, such as graphene, graphene oxide (GO), reduced GO (rGO), and functional GO (fGO). This process involves controlling multiple factors, such as graphene dispersion, viscosity, and post-curing, which impact the properties of the 3D-printed graphene-based constructs. To this end, those materials combined with 3D printing approaches have demonstrated prominent regeneration potential for bone, neural, cardiac, and skin tissues. Overall, graphene in 3D bioprinting may pave the way for new regenerative strategies with translational implications in orthopedics, neurology, and cardiovascular areas.
Collapse
Affiliation(s)
- Rahul Patil
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC 20057, USA;
- Center for Biological and Biomedical Engineering, Georgetown University, Washington, DC 20057, USA
| | - Stella Alimperti
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC 20057, USA;
- Center for Biological and Biomedical Engineering, Georgetown University, Washington, DC 20057, USA
| |
Collapse
|
43
|
Agarwal G, Roy A, Singh AA, Kumar H, Mandoli A, Srivastava A. BM-MSC-Loaded Graphene-Collagen Cryogels Ameliorate Neuroinflammation in a Rat Spinal Cord Injury Model. ACS APPLIED BIO MATERIALS 2024; 7:1478-1489. [PMID: 38354406 DOI: 10.1021/acsabm.3c00876] [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: 02/16/2024]
Abstract
A major obstacle to axonal regeneration following spinal cord injury (SCI) is neuroinflammation mediated by astrocytes and microglial cells. We previously demonstrated that graphene-based collagen hydrogels alone can decrease neuroinflammation in SCI. Their regenerative potential, however, is poorly understood and incomplete. Furthermore, stem cells have demonstrated both neuroprotective and regenerative properties in spinal cord regeneration, although there are constraints connected with the application of stem cell-based therapy. In this study, we have analyzed the regeneration capability of human bone marrow mesenchymal stem cell (BM-MSC)-loaded graphene-cross-linked collagen cryogels (Gr-Col) in a thoracic (T10-T11) hemisection model of SCI. Our study found that BM-MSC-loaded Gr-Col improves axonal regeneration, reduces neuroinflammation by decreasing astrocyte reactivity, and promotes M2 macrophage polarization. BM-MSC-loaded-Gr-Col demonstrated enhanced regenerative potential compared to Gr-Col and the injury group control. Next-generation sequencing (NGS) analysis revealed that BM-MSC-loaded-Gr-Col modulates the JAK2-STAT3 pathway, thus decreasing the reactive and scar-forming astrocyte phenotype. The decrease in neuroinflammation in the BM-MSC-loaded-Gr-Col group is attributed to the modulation of Notch/Rock and STAT5a/b and STAT6 signaling. Overall, Gene Set Enrichment Analysis suggests the promising role of BM-MSC-loaded-Gr-Col in promoting axonal regeneration after SCI by modulating molecular pathways such as the PI3/Akt pathway, focal adhesion kinase, and various inflammatory pathways.
Collapse
Affiliation(s)
- Gopal Agarwal
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Abhishek Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Abhishek A Singh
- Department of Molecular Biology, Radboud University, Postbus 9101, Nijmegen 6500 HB, The Netherlands
| | - Hemant Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Amit Mandoli
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Akshay Srivastava
- Department of Medical Device, National Institute of Pharmaceutical Education and Research, Ahmedabad, Gandhinagar, Gujarat 382355, India
| |
Collapse
|
44
|
Ferronato GDA, Vit FF, da Silveira JC. 3D culture applied to reproduction in females: possibilities and perspectives. Anim Reprod 2024; 21:e20230039. [PMID: 38510565 PMCID: PMC10954237 DOI: 10.1590/1984-3143-ar2023-0039] [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: 03/20/2023] [Accepted: 12/13/2023] [Indexed: 03/22/2024] Open
Abstract
In vitro cell culture is a well-established technique present in numerous laboratories in diverse areas. In reproduction, gametes, embryos, and reproductive tissues, such as the ovary and endometrium, can be cultured. These cultures are essential for embryo development studies, understanding signaling pathways, developing drugs for reproductive diseases, and in vitro embryo production (IVP). Although many culture systems are successful, they still have limitations to overcome. Three-dimensional (3D) culture systems can be close to physiological conditions, allowing greater interaction between cells and cells with the surrounding environment, maintenance of the cells' natural morphology, and expression of genes and proteins such as in vivo. Additionally, three-dimensional culture systems can stimulated extracellular matrix generating responses due to the mechanical force produced. Different techniques can be used to perform 3D culture systems, such as hydrogel matrix, hanging drop, low attachment surface, scaffold, levitation, liquid marble, and 3D printing. These systems demonstrate satisfactory results in follicle culture, allowing the culture from the pre-antral to antral phase, maintaining the follicular morphology, and increasing the development rates of embryos. Here, we review some of the different techniques of 3D culture systems and their applications to the culture of follicles and embryos, bringing new possibilities to the future of assisted reproduction.
Collapse
Affiliation(s)
| | - Franciele Flores Vit
- Faculdade de Zootecnia e Engenharia de Alimentos, Universidade de São Paulo, Pirassununga, SP, Brasil
| | | |
Collapse
|
45
|
Bai B, Liu Y, Huang J, Wang S, Chen H, Huo Y, Zhou H, Liu Y, Feng S, Zhou G, Hua Y. Tolerant and Rapid Endochondral Bone Regeneration Using Framework-Enhanced 3D Biomineralized Matrix Hydrogels. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305580. [PMID: 38127989 PMCID: PMC10916654 DOI: 10.1002/advs.202305580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 11/01/2023] [Indexed: 12/23/2023]
Abstract
Tissue-engineered bone has emerged as a promising alternative for bone defect repair due to the advantages of regenerative bone healing and physiological functional reconstruction. However, there is very limited breakthrough in achieving favorable bone regeneration due to the harsh osteogenic microenvironment after bone injury, especially the avascular and hypoxic conditions. Inspired by the bone developmental mode of endochondral ossification, a novel strategy is proposed for tolerant and rapid endochondral bone regeneration using framework-enhanced 3D biomineralized matrix hydrogels. First, it is meticulously designed 3D biomimetic hydrogels with both hypoxic and osteoinductive microenvironment, and then integrated 3D-printed polycaprolactone framework to improve their mechanical strength and structural fidelity. The inherent hypoxic 3D matrix microenvironment effectively activates bone marrow mesenchymal stem cells self-regulation for early-stage chondrogenesis via TGFβ/Smad signaling pathway due to the obstacle of aerobic respiration. Meanwhile, the strong biomineralized microenvironment, created by a hybrid formulation of native-constitute osteogenic inorganic salts, can synergistically regulate both bone mineralization and osteoclastic differentiation, and thus accelerate the late-stage bone maturation. Furthermore, both in vivo ectopic osteogenesis and in situ skull defect repair successfully verified the high efficiency and mechanical maintenance of endochondral bone regeneration mode, which offers a promising treatment for craniofacial bone defect repair.
Collapse
Affiliation(s)
- Baoshuai Bai
- Shanghai Key Laboratory of Tissue EngineeringDepartment of Plastic and Reconstructive Surgery of Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
- National Tissue Engineering Center of ChinaShanghai200241P. R. China
- Department of OrthopaedicsAdvanced Medical Research InstituteQilu Hospital of Shangdong University Centre for OrthopaedicsShandong UniversityJinanShandong250100P. R. China
- Department of OrthopaedicsCheeloo College of MedicineThe Second Hospital of Shandong UniversityShandong UniversityJinanShandong250033P. R. China
| | - Yanhan Liu
- Shanghai Key Laboratory of Tissue EngineeringDepartment of Plastic and Reconstructive Surgery of Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
- National Tissue Engineering Center of ChinaShanghai200241P. R. China
- Department of OphthalmologyRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200127P. R. China
| | - Jinyi Huang
- Shanghai Key Laboratory of Tissue EngineeringDepartment of Plastic and Reconstructive Surgery of Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
- National Tissue Engineering Center of ChinaShanghai200241P. R. China
| | - Sinan Wang
- Shanghai Key Laboratory of Tissue EngineeringDepartment of Plastic and Reconstructive Surgery of Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
- National Tissue Engineering Center of ChinaShanghai200241P. R. China
| | - Hongying Chen
- Shanghai Key Laboratory of Tissue EngineeringDepartment of Plastic and Reconstructive Surgery of Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
- National Tissue Engineering Center of ChinaShanghai200241P. R. China
| | - Yingying Huo
- Shanghai Key Laboratory of Tissue EngineeringDepartment of Plastic and Reconstructive Surgery of Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
- National Tissue Engineering Center of ChinaShanghai200241P. R. China
| | - Hengxing Zhou
- Department of OrthopaedicsAdvanced Medical Research InstituteQilu Hospital of Shangdong University Centre for OrthopaedicsShandong UniversityJinanShandong250100P. R. China
- Department of OrthopaedicsCheeloo College of MedicineThe Second Hospital of Shandong UniversityShandong UniversityJinanShandong250033P. R. China
| | - Yu Liu
- Shanghai Key Laboratory of Tissue EngineeringDepartment of Plastic and Reconstructive Surgery of Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
- National Tissue Engineering Center of ChinaShanghai200241P. R. China
| | - Shiqing Feng
- Department of OrthopaedicsAdvanced Medical Research InstituteQilu Hospital of Shangdong University Centre for OrthopaedicsShandong UniversityJinanShandong250100P. R. China
- Department of OrthopaedicsCheeloo College of MedicineThe Second Hospital of Shandong UniversityShandong UniversityJinanShandong250033P. R. China
| | - Guangdong Zhou
- Shanghai Key Laboratory of Tissue EngineeringDepartment of Plastic and Reconstructive Surgery of Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
- National Tissue Engineering Center of ChinaShanghai200241P. R. China
| | - Yujie Hua
- Shanghai Key Laboratory of Tissue EngineeringDepartment of Plastic and Reconstructive Surgery of Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghai200011P. R. China
- National Tissue Engineering Center of ChinaShanghai200241P. R. China
| |
Collapse
|
46
|
Lorion C, Bardin V, Bonnet S, Lopez-Gaydon A, Vogelgesang B, Bechetoille N. Elastogenic potential and antisagging properties of a novel Murraya koenigii extract. J Cosmet Dermatol 2024; 23:1036-1044. [PMID: 37997524 DOI: 10.1111/jocd.16059] [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: 07/11/2023] [Revised: 10/04/2023] [Accepted: 10/20/2023] [Indexed: 11/25/2023]
Abstract
BACKGROUND The process by which functional elastic fibers are produced, namely elastogenesis, is complex and difficult to assess in vitro. Identifying efficient elasticity-boosting ingredients thus represents a challenge. AIMS The elasticity-boosting properties of a novel extract of Murraya koenigii leafy stems were assessed in vitro in 3D culture models before being evaluated in human female volunteers. METHODS Synthesis of elastic fiber related proteins was evaluated in a skin-equivalent model. Using multiphoton microscopy, the structural organization of elastin deposits was studied within a scaffold-free dermal microtissue. Biomechanical properties of the 3D microtissue were also measured by atomic force microscopy. In vivo, fringe-projection and image analysis were used to evaluate nasogenian fold severity in a panel of Caucasian female volunteers. The impact of gravity on visible signs of facial aging was assessed by clinical scoring carried out alternatively in the supine and sitting positions. RESULTS We showed the Murraya koenigii extract increased protein expressions of elastin and fibrillin-1 in a 3D skin equivalent model. Using scaffold-free dermal microtissue, we confirmed that Murraya koenigii extract allowed a proper and ordered network of elastin deposits and consequently improved tissue elasticity. Clinical data showed that a twice-daily application for 98 days of the extract formulated at 1% allowed to visibly reduce nasogenian fold severity, jowl severity and to mitigate the impact of gravity on the facial signs of aging. CONCLUSION The newly discovered extract of Murraya koenigii leafy stems represents an innovative antiaging ingredient suited for elasticity-boosting and antisagging claims.
Collapse
|
47
|
Regato-Herbella M, Morhenn I, Mantione D, Pascuzzi G, Gallastegui A, Caribé dos Santos Valle AB, Moya SE, Criado-Gonzalez M, Mecerreyes D. ROS-Responsive 4D Printable Acrylic Thioether-Based Hydrogels for Smart Drug Release. CHEMISTRY OF MATERIALS : A PUBLICATION OF THE AMERICAN CHEMICAL SOCIETY 2024; 36:1262-1272. [PMID: 38370279 PMCID: PMC10870821 DOI: 10.1021/acs.chemmater.3c02264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 11/30/2023] [Accepted: 12/01/2023] [Indexed: 02/20/2024]
Abstract
Reactive oxygen species (ROS) play a key role in several biological functions like regulating cell survival and signaling; however, their effect can range from beneficial to nondesirable oxidative stress when they are overproduced causing inflammation or cancer diseases. Thus, the design of tailor-made ROS-responsive polymers offers the possibility of engineering hydrogels for target therapies. In this work, we developed thioether-based ROS-responsive difunctional monomers from ethylene glycol/thioether acrylate (EGnSA) with different lengths of the EGn chain (n = 1, 2, 3) by the thiol-Michael addition click reaction. The presence of acrylate groups allowed their photopolymerization by UV light, while the thioether groups conferred ROS-responsive properties. As a result, smart PEGnSA hydrogels were obtained, which could be processed by four-dimensional (4D) printing. The mechanical properties of the hydrogels were determined by rheology, pointing out a decrease of the elastic modulus (G') with the length of the EG segment. To enhance the stability of the hydrogels after swelling, the EGnSA monomers were copolymerized with a polar monomer, 2-hydroxyethyl acrylate (HEA), leading to P[(EGnSA)x-co-HEAy] with improved compatibility in aqueous media, making it a less brittle material. Swelling properties of the hydrogels increased in the presence of hydrogen peroxide, a kind of ROS, reaching values of ≈130% for P[(EG3SA)7-co-HEA93] which confirms the stimuli-responsive properties. Then, the P[(EG3SA)x-co-HEAy] hydrogels were employed as matrixes for the encapsulation of a chemotherapeutic drug, 5-fluorouracil (5FU), which showed sustained release over time modulated by the presence of H2O2. Finally, the effect of the 5-FU release from P[(EG3SA)x-co-HEAy] hydrogels was tested in vitro with melanoma cancer cells B16F10, pointing out B16F10 growth inhibition values in the range of 40-60% modulated by the EG3SA percentage and the presence or absence of ROS agents, thus confirming their excellent ROS-responsive properties for the treatment of localized pathologies.
Collapse
Affiliation(s)
- Maria Regato-Herbella
- POLYMAT
University of the Basque Country UPV/EHU, Joxe Mari Korta Center. Avda. Tolosa 72, 20018 Donostia-San Sebastián, Spain
- Center
for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo de Miramón 194, 20014Donostia-San Sebastián, Spain
| | - Isabel Morhenn
- POLYMAT
University of the Basque Country UPV/EHU, Joxe Mari Korta Center. Avda. Tolosa 72, 20018 Donostia-San Sebastián, Spain
| | - Daniele Mantione
- POLYMAT
University of the Basque Country UPV/EHU, Joxe Mari Korta Center. Avda. Tolosa 72, 20018 Donostia-San Sebastián, Spain
- Ikerbasque,
Basque Foundation for Science, 48013 Bilbao, Spain
| | - Giuseppe Pascuzzi
- Department
of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133 Milano ,Italy
| | - Antonela Gallastegui
- POLYMAT
University of the Basque Country UPV/EHU, Joxe Mari Korta Center. Avda. Tolosa 72, 20018 Donostia-San Sebastián, Spain
| | - Ana Beatriz Caribé dos Santos Valle
- Center
for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo de Miramón 194, 20014Donostia-San Sebastián, Spain
| | - Sergio E. Moya
- Center
for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo de Miramón 194, 20014Donostia-San Sebastián, Spain
| | - Miryam Criado-Gonzalez
- POLYMAT
University of the Basque Country UPV/EHU, Joxe Mari Korta Center. Avda. Tolosa 72, 20018 Donostia-San Sebastián, Spain
| | - David Mecerreyes
- POLYMAT
University of the Basque Country UPV/EHU, Joxe Mari Korta Center. Avda. Tolosa 72, 20018 Donostia-San Sebastián, Spain
- Ikerbasque,
Basque Foundation for Science, 48013 Bilbao, Spain
| |
Collapse
|
48
|
Yildiz SN, Sezgin Arslan T, Arslan YE. Organic-inorganic biohybrid films from wool-keratin/jellyfish-collagen/silica/boron via sol-gel reactions for soft tissue engineering applications. Biomed Mater 2024; 19:025032. [PMID: 38306684 DOI: 10.1088/1748-605x/ad2557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 02/02/2024] [Indexed: 02/04/2024]
Abstract
Therapeutic angiogenesis is pivotal in creating effective tissue-engineered constructs that deliver nutrients and oxygen to surrounding cells. Hence, biomaterials that promote angiogenesis can enhance the efficacy of various medical treatments, encompassing tissue engineering, wound healing, and drug delivery systems. Considering these, we propose a rapid method for producing composite silicon-boron-wool keratin/jellyfish collagen (Si-B-WK/JFC) inorganic-organic biohybrid films using sol-gel reactions. In this approach, reactive tetraethyl orthosilicate and boric acid (pKa ⩾ 9.24) were used as silicon and boron sources, respectively, and a solid-state gel was formed through the condensation reaction of these reactive groups with the keratin/collagen mixture. Once the resulting gel was thoroughly suspended in water, the films were prepared by a casting/solvent evaporation methodology. The fabricated hybrid films were characterized structurally and mechanically. In addition, angiogenic characteristics were determined by the in ovo chick chorioallantoic membrane assay, which revealed an increased vascular network within the Si-B-WK/JFC biohybrid films. In conclusion, it is believed that Si-B-WK/JFC biohybrid films with mechanical and pro-angiogenic properties have the potential to be possessed in soft tissue engineering applications, especially wound healing.
Collapse
Affiliation(s)
- Safiye Nur Yildiz
- Regenerative Biomaterials Laboratory, Department of Bioengineering, Faculty of Engineering, Canakkale Onsekiz Mart University, Canakkale 17100, Turkey
| | - Tugba Sezgin Arslan
- Regenerative Biomaterials Laboratory, Department of Bioengineering, Faculty of Engineering, Canakkale Onsekiz Mart University, Canakkale 17100, Turkey
| | - Yavuz Emre Arslan
- Regenerative Biomaterials Laboratory, Department of Bioengineering, Faculty of Engineering, Canakkale Onsekiz Mart University, Canakkale 17100, Turkey
| |
Collapse
|
49
|
Jiao X, Wu F, Yue X, Yang J, Zhang Y, Qiu J, Ke X, Sun X, Zhao L, Xu C, Li Y, Yang X, Yang G, Gou Z, Zhang L. New insight into biodegradable macropore filler on tuning mechanical properties and bone tissue ingrowth in sparingly dissolvable bioceramic scaffolds. Mater Today Bio 2024; 24:100936. [PMID: 38234459 PMCID: PMC10792586 DOI: 10.1016/j.mtbio.2023.100936] [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: 10/22/2023] [Revised: 12/21/2023] [Accepted: 12/27/2023] [Indexed: 01/19/2024] Open
Abstract
Structural parameters of the implants such as shape, size, and porosity of the pores have been extensively investigated to promote bone tissue repair, however, it is unknown how the pore interconnectivity affects the bone growth behaviors in the scaffolds. Herein we systematically evaluated the effect of biodegradable bioceramics as a secondary phase filler in the macroporous networks on the mechanical and osteogenic behaviors in sparingly dissolvable bioceramic scaffolds. The pure hardystonite (HT) scaffolds with ∼550 & 800 μm in pore sizes were prepared by digital light processing, and then the Sr-doped calcium silicate (SrCSi) bioceramic slurry without and with 30 % organic porogens were intruded into the HT scaffolds with 800 μm pore size and sintered at 1150 °C. It indicated that the organic porogens could endow spherical micropores in the SrCSi filler, and the invasion of the SrCSi component could not only significantly enhance the compressive strength and modulus of the HT-based scaffolds, but also induce osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs). The pure HT scaffolds showed extremely slow bio-dissolution in Tris buffer after immersion for 8 weeks (∼1 % mass decay); in contrast, the SrCSi filler would readily dissolve into the aqueous medium and produced a steady mass decay (>6 % mass loss). In vivo experiments in rabbit femoral bone defect models showed that the pure HT scaffolds showed bone tissue ingrowth but the bone growth was impeded in the SrCSi-intruded scaffolds within 4 weeks; however, the group with higher porosity of SrCSi filler showed appreciable osteogenesis after 8 weeks of implantation and the whole scaffold was uniformly covered by new bone tissues after 16 weeks. These findings provide some new insights that the pore interconnectivity is not inevitable to impede bone ingrowth with the prolongation of implantation time, and such a highly biodegradable and bioactive filler intrusion strategy may be beneficial for optimizing the performances of scaffolds in bone regenerative medicine applications.
Collapse
Affiliation(s)
- Xiaoyi Jiao
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Department of Orthopaedics, The Third Hospital Affiliated to Wenzhou Medical University & Rui'an People's Hospital, Rui'an, 325200, China
| | - Fanghui Wu
- Department of Orthopaedics, The Third Hospital Affiliated to Wenzhou Medical University & Rui'an People's Hospital, Rui'an, 325200, China
| | - Xusong Yue
- Department of Orthopaedics, The Third Hospital Affiliated to Wenzhou Medical University & Rui'an People's Hospital, Rui'an, 325200, China
| | - Jun Yang
- Department of Orthopaedics, The Third Hospital Affiliated to Wenzhou Medical University & Rui'an People's Hospital, Rui'an, 325200, China
| | - Yan Zhang
- Bio-nanomaterials and Regenerative Medicine Research Division, Zhejiang-California International Nanosystem Institute, Zhejiang University, Hangzhou, 310058, China
| | - Jiandi Qiu
- Department of Orthopaedics, The Third Hospital Affiliated to Wenzhou Medical University & Rui'an People's Hospital, Rui'an, 325200, China
| | - Xiurong Ke
- Department of Orthopaedics, The Third Hospital Affiliated to Wenzhou Medical University & Rui'an People's Hospital, Rui'an, 325200, China
| | - Xiaoliang Sun
- Department of Orthopaedics, The Third Hospital Affiliated to Wenzhou Medical University & Rui'an People's Hospital, Rui'an, 325200, China
| | - Liben Zhao
- Department of Orthopaedics, The Third Hospital Affiliated to Wenzhou Medical University & Rui'an People's Hospital, Rui'an, 325200, China
| | - Chuchu Xu
- Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Yifan Li
- Department of Orthopaedics, The First Affiliated Hospital, School of Medicine of Zhejiang University, Hangzhou, 310003, China
| | - Xianyan Yang
- Bio-nanomaterials and Regenerative Medicine Research Division, Zhejiang-California International Nanosystem Institute, Zhejiang University, Hangzhou, 310058, China
| | - Guojing Yang
- Department of Orthopaedics, The Third Hospital Affiliated to Wenzhou Medical University & Rui'an People's Hospital, Rui'an, 325200, China
| | - Zhongru Gou
- Bio-nanomaterials and Regenerative Medicine Research Division, Zhejiang-California International Nanosystem Institute, Zhejiang University, Hangzhou, 310058, China
| | - Lei Zhang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Department of Orthopaedics, The Third Hospital Affiliated to Wenzhou Medical University & Rui'an People's Hospital, Rui'an, 325200, China
| |
Collapse
|
50
|
Moreno Florez AI, Malagon S, Ocampo S, Leal-Marin S, Ossa EA, Glasmacher B, Garcia C, Pelaez-Vargas A. In vitro evaluation of the osteogenic and antimicrobial potential of porous wollastonite scaffolds impregnated with ethanolic extracts of propolis. Front Bioeng Biotechnol 2024; 12:1321466. [PMID: 38361789 PMCID: PMC10867276 DOI: 10.3389/fbioe.2024.1321466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Accepted: 01/11/2024] [Indexed: 02/17/2024] Open
Abstract
Context: The development of porous devices using materials modified with various natural agents has become a priority for bone healing processes in the oral and maxillofacial field. There must be a balance between the proliferation of eukaryotic and the inhibition of prokaryotic cells to achieve proper bone health. Infections might inhibit the formation of new alveolar bone during bone graft augmentation. Objective: This study aimed to evaluate the in vitro osteogenic behavior of human bone marrow stem cells and assess the antimicrobial response to 3D-printed porous scaffolds using propolis-modified wollastonite. Methodology: A fractional factorial design of experiments was used to obtain a 3D printing paste for developing scaffolds with a triply periodic minimal surface (TPMS) gyroid geometry based on wollastonite and modified with an ethanolic propolis extract. The antioxidant activity of the extracts was characterized using free radical scavenging methods (DPPH and ABTS). Cell proliferation and osteogenic potential using Human Bone Marrow Stem Cells (bmMSCs) were assessed at different culture time points up to 28 days. MIC and inhibition zones were studied from single strain cultures, and biofilm formation was evaluated on the scaffolds under co-culture conditions. The mechanical strength of the scaffolds was evaluated. Results: Through statistical design of experiments, a paste suitable for printing scaffolds with the desired geometry was obtained. Propolis extracts modifying the TPMS gyroid scaffolds showed favorable cell proliferation and metabolic activity with osteogenic potential after 21 days. Additionally, propolis exhibited antioxidant activity, which may be related to the antimicrobial effectiveness of the scaffolds against S. aureus and S. epidermidis cultures. The mechanical properties of the scaffolds were not affected by propolis impregnation. Conclusion: These results demonstrate that propolis-impregnated porous wollastonite scaffolds might have the potential to stimulate bone repair in maxillofacial tissue engineering applications.
Collapse
Affiliation(s)
- Ana Isabel Moreno Florez
- Grupo de Materiales Cerámicos y Vítreos, Universidad Nacional de Colombia Sede Medellín, Medellín, Colombia
| | - Sarita Malagon
- Grupo GIOM, Facultad de Odontología, Universidad Cooperativa de Colombia, Sede Medellín, Colombia
| | - Sebastian Ocampo
- Grupo de Materiales Cerámicos y Vítreos, Universidad Nacional de Colombia Sede Medellín, Medellín, Colombia
| | - Sara Leal-Marin
- Institute for Multiphase Processes (IMP), Leibniz University Hannover, Garbsen, Germany
- Lower Saxony Center for Biomedical Engineering, Implant Research and Development, Hannover, Germany
| | - Edgar Alexander Ossa
- School of Applied Sciences and Engineering, Universidad Eafit, Medellín, Colombia
| | - Birgit Glasmacher
- Institute for Multiphase Processes (IMP), Leibniz University Hannover, Garbsen, Germany
- Lower Saxony Center for Biomedical Engineering, Implant Research and Development, Hannover, Germany
| | - Claudia Garcia
- Grupo de Materiales Cerámicos y Vítreos, Universidad Nacional de Colombia Sede Medellín, Medellín, Colombia
| | - Alejandro Pelaez-Vargas
- Grupo GIOM, Facultad de Odontología, Universidad Cooperativa de Colombia, Sede Medellín, Colombia
| |
Collapse
|