1
|
Na KH, Lee HJ, Kim JH, Uddin MS, Park YH, Song YM, Park CS, Park JB. Enhancement of Chondrogenic Differentiation in Bone Marrow-Derived Stem Cell Spheroids by Cuminum cyminum Methanolic Extract: Insights into Concentration-Dependent mRNA Expression and Gene Clustering Analysis. J Pers Med 2024; 14:1142. [PMID: 39728055 PMCID: PMC11679761 DOI: 10.3390/jpm14121142] [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: 10/06/2024] [Revised: 12/02/2024] [Accepted: 12/03/2024] [Indexed: 12/28/2024] Open
Abstract
Background/Objectives: Cuminum cyminum L. has been utilized as a medicinal plant for centuries. This research sought to examine the effects of cumin methanolic extract (CMT) on the chondrogenic differentiation of human bone marrow-derived mesenchymal stem cells. Methods: Spheroids were generated using human stem cells and cultured with CMT at concentrations between 0 and 1 µg/mL. Morphological assessments and cell viability tests were conducted on days 1 and 3. Chondrogenic differentiation expression was evaluated through quantitative polymerase chain reaction, Western blot, and RNA sequencing. SOX9, FAM20B, COL2A1, and COL1A1 mRNA expression levels were determined using real-time polymerase chain reaction. Protein expression was analyzed via Western blot. Results: Throughout this study, the spheroids maintained their integrity and shape. No significant variations in spheroid diameter were observed among the groups. CMT treatment enhanced the expression of SOX9 and FAM20B. Conclusions: The methanolic extract of Cuminum cyminum facilitated chondrogenic differentiation in human bone marrow-derived mesenchymal stem cells by modulating SOX9 and FAM20B expression. This indicates its potential application in cartilage tissue engineering.
Collapse
Affiliation(s)
- Kyung-Hwan Na
- Department of Medicine, Graduate School, The Catholic University of Korea, Seoul 06591, Republic of Korea;
| | - Hyun-Jin Lee
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (H.-J.L.); (J.-H.K.); (Y.-M.S.)
| | - Ju-Hwan Kim
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (H.-J.L.); (J.-H.K.); (Y.-M.S.)
| | - Md. Salah Uddin
- Ethnobotanical Database of Bangladesh, Tejgaon, Dhaka 1208, Bangladesh;
| | | | - Young-Min Song
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (H.-J.L.); (J.-H.K.); (Y.-M.S.)
| | - Chul-Sung Park
- Department of Biomedical Science, Graduate School of Biomedical Science & Engineering, Hanyang University, Seoul 04763, Republic of Korea;
| | - Jun-Beom Park
- Department of Medicine, Graduate School, The Catholic University of Korea, Seoul 06591, Republic of Korea;
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (H.-J.L.); (J.-H.K.); (Y.-M.S.)
- Dental Implantology, Graduate School of Clinical Dental Science, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
2
|
Wang M, Wang J, Xu X, Li E, Xu P. Engineering gene-activated bioprinted scaffolds for enhancing articular cartilage repair. Mater Today Bio 2024; 29:101351. [PMID: 39649247 PMCID: PMC11621797 DOI: 10.1016/j.mtbio.2024.101351] [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: 09/29/2024] [Revised: 11/14/2024] [Accepted: 11/18/2024] [Indexed: 12/10/2024] Open
Abstract
Untreated articular cartilage injuries often result in severe chronic pain and dyskinesia. Current repair strategies have limitations in effectively promoting articular cartilage repair, underscoring the need for innovative therapeutic approaches. A gene-activated matrix (GAM) is a promising and comprehensive therapeutic strategy that integrates tissue-engineered scaffold-guided gene therapy to promote long-term articular cartilage repair by enhancing gene retention, reducing gene loss, and regulating gene release. However, for effective articular cartilage repair, the GAM scaffold must mimic the complex gradient structure of natural articular cartilage. Three-dimensional (3D) bioprinting technology has emerged as a compelling solution, offering the ability to precisely create complex microstructures that mimic the natural articular cartilage. In this review, we summarize the recent research progress on GAM and 3D bioprinted scaffolds in articular cartilage tissue engineering (CTE), while also exploring future challenges and development directions. This review aims to provide new ideas and concepts for the development of gene-activated bioprinted scaffolds with specific properties tailored to meet the stringent requirements of articular cartilage repair.
Collapse
Affiliation(s)
- Min Wang
- Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710000, China
- Xi'an Key Laboratory of Pathogenesis and Precision Treatment of Arthritis, Xi'an, 710000, China
| | - Jiachen Wang
- Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710000, China
- Xi'an Key Laboratory of Pathogenesis and Precision Treatment of Arthritis, Xi'an, 710000, China
| | - Xin Xu
- Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710000, China
- Xi'an Key Laboratory of Pathogenesis and Precision Treatment of Arthritis, Xi'an, 710000, China
| | - Erliang Li
- Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710000, China
- Xi'an Key Laboratory of Pathogenesis and Precision Treatment of Arthritis, Xi'an, 710000, China
| | - Peng Xu
- Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710000, China
- Xi'an Key Laboratory of Pathogenesis and Precision Treatment of Arthritis, Xi'an, 710000, China
| |
Collapse
|
3
|
Cheng L, Zhou Z, Li Q, Li W, Li X, Li G, Fan J, Yu L, Yin G. Dendronized chitosan hydrogel with GIT1 to accelerate bone defect repair through increasing local neovascular amount. Bone Rep 2023; 19:101712. [PMID: 37744736 PMCID: PMC10511783 DOI: 10.1016/j.bonr.2023.101712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 07/15/2023] [Accepted: 08/29/2023] [Indexed: 09/26/2023] Open
Abstract
Bone defects have long been a major healthcare issue because of the difficulties in regenerating bone mass volume and the high cost of treatment. G protein-coupled receptor kinase 2 interacting protein 1 (GIT1) has been proven to play an important role both in vascular development and in bone fracture healing. In this study, a type of thermoresponsive injectable hydrogel from oligoethylene glycol-based dendronized chitosan (G1-CS) was loaded with GIT1-plasmids (G1-CS/GIT1), and used to fill unicortical bone defects. RT-PCR analysis confirmed that G1-CS/GIT1 enhanced DNA transfection in MSCs both in vitro and in vivo. From the results of micro-CT, RT-PCR and histological analysis, it can be concluded that G1-CS/GIT1 accelerated the bone healing rate and increased the amount of neovascularization around the bone defects. In addition, an adeno-associated virus (AAV)-GIT1 was constructed to transfect mesenchymal stem cells. The results of capillary tube formation assay, immunofluorescence staining and western blot analysis proved that high expression of GIT1 induces mesenchymal stem cells to differentiate into endothelial cells. RT-PCR analysis and capillary tube formation assay confirmed that the Notch signaling pathway was activated in the differentiation process. Overall, we developed an efficient strategy through combination of injectable hydrogel and G1T1 for bone tissue engineering.
Collapse
Affiliation(s)
- Lin Cheng
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Huaihai West Road 99, Xuzhou, Jiangsu Province 221000, China
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, Jiangsu Province 210000, China
| | - Zhimin Zhou
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, Jiangsu Province 210000, China
| | - Qingqing Li
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, Jiangsu Province 210000, China
| | - Wen Li
- School of Materials Science and Engineering, Shanghai University, Nanchen Street 333, Shanghai 200444, China
| | - Xin Li
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Huaihai West Road 99, Xuzhou, Jiangsu Province 221000, China
| | - Gen Li
- Department of Orthopedics, The Affiliated Hospital of Xuzhou Medical University, Huaihai West Road 99, Xuzhou, Jiangsu Province 221000, China
| | - Jin Fan
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, Jiangsu Province 210000, China
| | - Lipeng Yu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, Jiangsu Province 210000, China
| | - Guoyong Yin
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, Jiangsu Province 210000, China
| |
Collapse
|
4
|
Anisiei A, Andreica BI, Mititelu-Tartau L, Coman CG, Bilyy R, Bila G, Rosca I, Sandu AI, Amler E, Marin L. Biodegradable trimethyl chitosan nanofiber mats by electrospinning as bioabsorbable dressings for wound closure and healing. Int J Biol Macromol 2023; 249:126056. [PMID: 37524280 DOI: 10.1016/j.ijbiomac.2023.126056] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 07/20/2023] [Accepted: 07/27/2023] [Indexed: 08/02/2023]
Abstract
The paper aimed to prepare quaternary chitosan-based nanofibers as bioabsorbable wound dressings. To this aim, fully biodegradable chitosan/N,N,N-trimethyl chitosan (TMC) nanofibers were designed and prepared via electrospinning, using poly(ethylene glycol) as sacrificial additive. The new biomaterials were structurally and morphologically characterized by FTIR and NMR spectroscopy, thermogravimetric analysis, X-ray diffraction and scanning electron microscopy, and their properties required for wound dressings application were investigated and discussed in detail. Thus, the nanofiber behavior was investigated by swelling, dynamic vapor sorption, and in vitro biodegradation in media mimicking the wound exudate. The mechanical properties were analysed from the stress-strain curves, the bioadhesivity from the texture analysis and the mucoadhesivity from the Zeta potential and transmittance measurements. The antimicrobial activity was assessed against S. aureus and E. coli strains, and the biocompatibility was tested in vitro on normal human dermal fibroblasts, and in vivo on rats. The application of the fiber mats with the best balance of properties as dressings on deep burn wound models in rats showed wound closure and active healing, with fully restoration of epithelia. It was concluded that the combination of chitosan with TMC into nanofibers provides new potential bioabsorbable wound dressing, opening new perspectives in regenerative medicine.
Collapse
Affiliation(s)
- Alexandru Anisiei
- "Petru Poni" Institute of Macromolecular Chemistry of Romanian Academy, Iasi, Romania
| | | | | | - Corneliu G Coman
- "Grigore T. Popa" University of Medicine and Pharmacy, Iasi, Romania
| | - Rostyslav Bilyy
- Lectinotest R&D, Mechamichna Str 2, 79022, Ukraine; Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
| | - Galyna Bila
- Lectinotest R&D, Mechamichna Str 2, 79022, Ukraine; Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
| | - Irina Rosca
- "Petru Poni" Institute of Macromolecular Chemistry of Romanian Academy, Iasi, Romania
| | - Andreea-Isabela Sandu
- "Petru Poni" Institute of Macromolecular Chemistry of Romanian Academy, Iasi, Romania
| | - Evžen Amler
- Research and Development Department Inocure, Prague, Czech Republic; Charles University, Prague, Czech Republic
| | - Luminita Marin
- "Petru Poni" Institute of Macromolecular Chemistry of Romanian Academy, Iasi, Romania.
| |
Collapse
|
5
|
Rasouli R, Paun RA, Tabrizian M. Sonoprinting nanoparticles on cellular spheroids via surface acoustic waves for enhanced nanotherapeutics delivery. LAB ON A CHIP 2023; 23:2091-2105. [PMID: 36942710 DOI: 10.1039/d2lc00854h] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Nanotherapeutics, on their path to the target tissues, face numerous physicochemical hindrances that affect their therapeutic efficacy. Physical barriers become more pronounced in pathological tissues, such as solid tumors, where they limit the penetration of nanocarriers into deeper regions, thereby preventing the efficient delivery of drug cargo. To address this challenge, we introduce a novel approach that employs surface acoustic wave (SAW) technology to sonoprint and enhance the delivery of nanoparticles onto and into cell spheroids. Our SAW platform is designed to generate focused and unidirectional acoustic waves for creating vigorous acoustic streaming while promoting Bjerknes forces. The effect of SAW excitation on cell viability, as well as the accumulation and penetration of nanoparticles on human breast cancer (MCF 7) and mouse melanoma (YUMM 1.7) cell spheroids were investigated. The high frequency, low input voltage, and contact-free nature of the proposed SAW system ensured over 92% cell viability for both cell lines after SAW exposure. SAW sonoprinting enhanced the accumulation of 100 nm polystyrene particles on the periphery of the spheroids to near four-fold, while the penetration of nanoparticles into the core regions of the spheroids was improved up to three times. To demonstrate the effectiveness of our SAW platform on the efficacy of nanotherapeutics, the platform was used to deliver nanoliposomes encapsulated with the anti-cancer metal compound copper diethyldithiocarbamate (CuET) to MCF 7 and YUMM 1.7 cell spheroids. A three-fold increase in the cytotoxic activity of the drug was observed in spheroids under the effect of SAW, compared to controls. The capacity of SAW-based devices to be manufactured as minuscule wearable patches can offer highly controllable, localized, and continuous acoustic waves to enhance drug delivery efficiency to target tissues.
Collapse
Affiliation(s)
- Reza Rasouli
- Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada.
| | - Radu Alexandru Paun
- Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada.
| | - Maryam Tabrizian
- Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada.
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada
| |
Collapse
|
6
|
Kim W, Gwon Y, Park S, Kim H, Kim J. Therapeutic strategies of three-dimensional stem cell spheroids and organoids for tissue repair and regeneration. Bioact Mater 2023; 19:50-74. [PMID: 35441116 PMCID: PMC8987319 DOI: 10.1016/j.bioactmat.2022.03.039] [Citation(s) in RCA: 86] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 03/16/2022] [Accepted: 03/25/2022] [Indexed: 02/07/2023] Open
Abstract
Three-dimensional (3D) stem cell culture systems have attracted considerable attention as a way to better mimic the complex interactions between individual cells and the extracellular matrix (ECM) that occur in vivo. Moreover, 3D cell culture systems have unique properties that help guide specific functions, growth, and processes of stem cells (e.g., embryogenesis, morphogenesis, and organogenesis). Thus, 3D stem cell culture systems that mimic in vivo environments enable basic research about various tissues and organs. In this review, we focus on the advanced therapeutic applications of stem cell-based 3D culture systems generated using different engineering techniques. Specifically, we summarize the historical advancements of 3D cell culture systems and discuss the therapeutic applications of stem cell-based spheroids and organoids, including engineering techniques for tissue repair and regeneration.
Collapse
Affiliation(s)
- Woochan Kim
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
- Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Yonghyun Gwon
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
- Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Sunho Park
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
- Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Hyoseong Kim
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
- Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Jangho Kim
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
- Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, 61186, Republic of Korea
| |
Collapse
|
7
|
Lee SY, Lee JW. 3D Spheroid Cultures of Stem Cells and Exosome Applications for Cartilage Repair. LIFE (BASEL, SWITZERLAND) 2022; 12:life12070939. [PMID: 35888029 PMCID: PMC9317836 DOI: 10.3390/life12070939] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/20/2022] [Accepted: 06/21/2022] [Indexed: 11/16/2022]
Abstract
Cartilage is a connective tissue that constitutes the structure of the body and consists of chondrocytes that produce considerable collagenous extracellular matrix and plentiful ground substances, such as proteoglycan and elastin fibers. Self-repair is difficult when the cartilage is damaged because of insufficient blood supply, low cellularity, and limited progenitor cell numbers. Therefore, three-dimensional (3D) culture systems, including pellet culture, hanging droplets, liquid overlays, self-injury, and spinner culture, have attracted attention. In particular, 3D spheroid culture strategies can enhance the yield of exosome production of mesenchymal stem cells (MSCs) when compared to two-dimensional culture, and can improve cellular restorative function by enhancing the paracrine effects of MSCs. Exosomes are membrane-bound extracellular vesicles, which are intercellular communication systems that carry RNAs and proteins. Information transfer affects the phenotype of recipient cells. MSC-derived exosomes can facilitate cartilage repair by promoting chondrogenic differentiation and proliferation. In this article, we reviewed recent major advances in the application of 3D culture techniques, cartilage regeneration with stem cells using 3D spheroid culture system, the effect of exosomes on chondrogenic differentiation, and chondrogenic-specific markers related to stem cell derived exosomes. Furthermore, the utilization of MSC-derived exosomes to enhance chondrogenic differentiation for osteoarthritis is discussed. If more mechanistic studies at the molecular level are conducted, MSC-spheroid-derived exosomes will supply a better therapeutic option to improve osteoarthritis.
Collapse
Affiliation(s)
- Seung Yeon Lee
- Department of Molecular Medicine, College of Medicine, Gachon University, 155, Gaetbeol-ro, Yeonsu-ku, Incheon 21999, Korea;
| | - Jin Woo Lee
- Department of Molecular Medicine, College of Medicine, Gachon University, 155, Gaetbeol-ro, Yeonsu-ku, Incheon 21999, Korea;
- Department of Health Sciences and Technology, GAIHST, Gachon University, 155, Gaetbeol-ro, Yeonsu-ku, Incheon 21999, Korea
- Correspondence: ; Tel.: +82-32-899-6516; Fax: +82-32-899-6039
| |
Collapse
|
8
|
Sun W, Zhang J, Qin Y, Tang H, Chen Y, Lin W, She Y, Zhang K, Yin J, Chen C. A Simple and Efficient Strategy for Preparing a Cell-Spheroid-Based Bioink. Adv Healthc Mater 2022; 11:e2200648. [PMID: 35543489 DOI: 10.1002/adhm.202200648] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/19/2022] [Indexed: 12/28/2022]
Abstract
Cell spheroids are a promising bioprinting building block that can mimic several physiological conditions in embryonic development. However, it remains challenging to efficiently prepare cell-spheroid-based bioink (Sph-bioink) with favorable printability and spheroid fusion ability. In this work, a poly(N-isopropylacrylamide) (PNIPAAm)-based porous hydrogel is developed as an "all-in-one" platform for Sph-bioink preparation. On the one hand, the nonadhesive porous structure in hydrogels is an effective tool for fabricating adipose-derived stem cell (ASC) spheroids in high yield, and the hydrogel itself also serves as a "carrier" for conveniently transferring cell spheroids to the bioprinter. On the other hand, the integration of redox/thermo-responsiveness allows the hydrogel to shift from a solid spheroid-making tool to an extrudable bioprinting medium that is sensitive to temperature. These features enabled a simple procedure for preparing Sph-bioink, in which the cell spheroids were densely packed to retain fusion capability. The present study also demonstrates that ASC spheroids formed in hydrogels have good biological preservation and superior chondrogenic differentiation, and verified the feasibility of using Sph-bioink to build custom-shaped mature cartilage. In conclusion, this strategy provides a simple, efficient, and standardized approach for Sph-bioink preparation, making it possible to produce tissue-engineered constructs with accelerated maturation and functionalization.
Collapse
Affiliation(s)
- Weiyan Sun
- Department of Thoracic Surgery Shanghai Pulmonary Hospital Tongji University School of Medicine Shanghai 200433 P. R. China
| | - Jiahui Zhang
- Department of Polymer Materials School of Materials Science and Engineering Shanghai University Shanghai 200444 P. R. China
| | - Yechi Qin
- Department of Polymer Materials School of Materials Science and Engineering Shanghai University Shanghai 200444 P. R. China
| | - Hai Tang
- Department of Thoracic Surgery Shanghai Pulmonary Hospital Tongji University School of Medicine Shanghai 200433 P. R. China
| | - Yi Chen
- Department of Thoracic Surgery Shanghai Pulmonary Hospital Tongji University School of Medicine Shanghai 200433 P. R. China
| | - Weikang Lin
- Department of Thoracic Surgery Shanghai Pulmonary Hospital Tongji University School of Medicine Shanghai 200433 P. R. China
| | - Yunlang She
- Department of Thoracic Surgery Shanghai Pulmonary Hospital Tongji University School of Medicine Shanghai 200433 P. R. China
- Shanghai Engineering Research Center of Lung Transplantation Shanghai 200433 P. R. China
| | - Kunxi Zhang
- Department of Polymer Materials School of Materials Science and Engineering Shanghai University Shanghai 200444 P. R. China
- Interventional Cancer Institute of Chinese Integrative Medicine Putuo Hospital Shanghai University of Traditional Chinese Medicine Shanghai 200060 P. R. China
| | - Jingbo Yin
- Department of Polymer Materials School of Materials Science and Engineering Shanghai University Shanghai 200444 P. R. China
| | - Chang Chen
- Department of Thoracic Surgery Shanghai Pulmonary Hospital Tongji University School of Medicine Shanghai 200433 P. R. China
- Shanghai Engineering Research Center of Lung Transplantation Shanghai 200433 P. R. China
| |
Collapse
|
9
|
Development of a fibrin-mediated gene delivery system for the treatment of cystinosis via design of experiment. Sci Rep 2022; 12:3752. [PMID: 35260693 PMCID: PMC8904479 DOI: 10.1038/s41598-022-07750-y] [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: 11/08/2021] [Accepted: 02/11/2022] [Indexed: 11/23/2022] Open
Abstract
Cystinosis is a rare disease, caused by a mutation in the gene cystinosin and characterised by the accumulation of cystine crystals. Advantages of biomaterial-mediated gene delivery include reduced safety concerns and the possibility to cure organs that are difficult to treat using systemic gene transfer methods. This study developed novel fibrin hydrogels for controlled, localised gene delivery, for the treatment of cystinosis. In the first part, fabrication parameters (i.e., DNA, thrombin, and aprotinin concentrations) were optimised, using a Design of Experiment (DOE) methodology. DOE is a statistical engineering approach to process optimisation, which increases experimental efficiency, reduces the number of experiments, takes into consideration interactions between different parameters, and allows the creation of predictive models. This study demonstrated the utility of DOE to the development of gene delivery constructs. In the second part of the study, primary fibroblasts from a patient with cystinosis were seeded on the biomaterials. Seeded cells expressed the recombinant CTNS and showed a decrease in cystine content. Furthermore, conditioned media contained functional copies of the recombinant CTNS. These were taken up by monolayer cultures of non-transfected cells. This study described a methodology to develop gene delivery constructs by using a DOE approach and ultimately provided new insights into the treatment of cystinosis.
Collapse
|
10
|
He W, Wang Q, Tian X, Pan G. Recapitulating dynamic ECM ligand presentation at biomaterial interfaces: Molecular strategies and biomedical prospects. EXPLORATION (BEIJING, CHINA) 2022; 2:20210093. [PMID: 37324582 PMCID: PMC10191035 DOI: 10.1002/exp.20210093] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 11/29/2021] [Indexed: 06/14/2023]
Abstract
The extracellular matrix (ECM) provides not only physical support for the tissue structural integrity, but also dynamic biochemical cues capable of regulating diverse cell behaviors and functions. Biomaterial surfaces with dynamic ligand presentation are capable of mimicking the dynamic biochemical cues of ECM, showing ECM-like functions to modulate cell behaviors. This review paper described an overview of present dynamic biomaterial interfaces by focusing on currently developed molecular strategies for dynamic ligand presentation. The paradigmatic examples for each strategy were separately discussed. In addition, the regulation of some typical cell behaviors on these dynamic biointerfaces including cell adhesion, macrophage polarization, and stem cell differentiation, and their potential applications in pathogenic cell isolation, single cell analysis, and tissue engineering are highlighted. We hope it would not only clarify a clear background of this field, but also inspire to exploit novel molecular strategies and more applications to match the increasing demand of manipulating complex cellular processes in biomedicine.
Collapse
Affiliation(s)
- Wenbo He
- Institute for Advanced MaterialsSchool of Materials Science and EngineeringJiangsu UniversityZhenjiangP. R. China
| | - Qinghe Wang
- Institute for Advanced MaterialsSchool of Materials Science and EngineeringJiangsu UniversityZhenjiangP. R. China
| | - Xiaohua Tian
- Institute for Advanced MaterialsSchool of Materials Science and EngineeringJiangsu UniversityZhenjiangP. R. China
- School of Chemistry and Chemical EngineeringJiangsu UniversityZhenjiangP. R. China
| | - Guoqing Pan
- Institute for Advanced MaterialsSchool of Materials Science and EngineeringJiangsu UniversityZhenjiangP. R. China
| |
Collapse
|
11
|
Lee J, Lee S, Huh SJ, Kang BJ, Shin H. Directed Regeneration of Osteochondral Tissue by Hierarchical Assembly of Spatially Organized Composite Spheroids. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103525. [PMID: 34806336 PMCID: PMC8787388 DOI: 10.1002/advs.202103525] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 10/20/2021] [Indexed: 05/11/2023]
Abstract
The use of engineered scaffolds or stem cells is investigated widely in the repair of injured musculoskeletal tissue. However, the combined regeneration of hierarchical osteochondral tissue remains a challenge due to delamination between cartilage and subchondral bone or difficulty in spatial control over differentiation of transplanted stem cells. Here, two types of composite spheroids are prepared using adipose-derived stem cells (hADSCs) and nanofibers coated with either transforming growth factor-β3 or bone morphogenetic growth factor-2 for chondrogenesis or osteogenesis, respectively. Each type of spheroid is then cultured within a 3D-printed microchamber in a spatially arranged manner to recapitulate the bilayer structure of osteochondral tissue. The presence of inductive factors regionally modulates in vitro chondrogenic or osteogenic differentiation of hADSCs within the biphasic construct without dedifferentiation. Furthermore, hADSCs from each spheroid proliferate and sprout and successfully connect the two layers mimicking the osteochondral interface without apertures. In vivo transplantation of the biphasic construct onto a femoral trochlear groove defect in rabbit knee joint results in 21.2 ± 2.8% subchondral bone volume/total volume and a cartilage score of 25.0 ± 3.7. The present approach can be an effective therapeutic platform to engineer complex tissue.
Collapse
Affiliation(s)
- Jinkyu Lee
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 04763, Republic of Korea
- BK21 FOUR Human-Tech Convergence Program, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 04763, Republic of Korea
| | - Seoyun Lee
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine and Research Institute for Veterinary Science, BK21 FOUR Future Veterinary Medicine Leading Education and Research Center, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Seung Jae Huh
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 04763, Republic of Korea
- BK21 FOUR Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 04763, Republic of Korea
| | - Byung-Jae Kang
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine and Research Institute for Veterinary Science, BK21 FOUR Future Veterinary Medicine Leading Education and Research Center, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Heungsoo Shin
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 04763, Republic of Korea
- BK21 FOUR Education and Research Group for Biopharmaceutical Innovation Leader, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 04763, Republic of Korea
- Institute of Nano Science and Technology, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 04763, Republic of Korea
| |
Collapse
|
12
|
Graceffa V. Physical and mechanical cues affecting biomaterial-mediated plasmid DNA delivery: insights into non-viral delivery systems. J Genet Eng Biotechnol 2021; 19:90. [PMID: 34142237 PMCID: PMC8211807 DOI: 10.1186/s43141-021-00194-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 06/09/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND Whilst traditional strategies to increase transfection efficiency of non-viral systems aimed at modifying the vector or the polyplexes/lipoplexes, biomaterial-mediated gene delivery has recently sparked increased interest. This review aims at discussing biomaterial properties and unravelling underlying mechanisms of action, for biomaterial-mediated gene delivery. DNA internalisation and cytoplasmic transport are initially discussed. DNA immobilisation, encapsulation and surface-mediated gene delivery (SMD), the role of extracellular matrix (ECM) and topographical cues, biomaterial stiffness and mechanical stimulation are finally outlined. MAIN TEXT Endocytic pathways and mechanisms to escape the lysosomal network are highly variable. They depend on cell and DNA complex types but can be diverted using appropriate biomaterials. 3D scaffolds are generally fabricated via DNA immobilisation or encapsulation. Degradation rate and interaction with the vector affect temporal patterns of DNA release and transgene expression. In SMD, DNA is instead coated on 2D surfaces. SMD allows the incorporation of topographical cues, which, by inducing cytoskeletal re-arrangements, modulate DNA endocytosis. Incorporation of ECM mimetics allows cell type-specific transfection, whereas in spite of discordances in terms of optimal loading regimens, it is recognised that mechanical loading facilitates gene transfection. Finally, stiffer 2D substrates enhance DNA internalisation, whereas in 3D scaffolds, the role of stiffness is still dubious. CONCLUSION Although it is recognised that biomaterials allow the creation of tailored non-viral gene delivery systems, there still are many outstanding questions. A better characterisation of endocytic pathways would allow the diversion of cell adhesion processes and cytoskeletal dynamics, in order to increase cellular transfection. Further research on optimal biomaterial mechanical properties, cell ligand density and loading regimens is limited by the fact that such parameters influence a plethora of other different processes (e.g. cellular adhesion, spreading, migration, infiltration, and proliferation, DNA diffusion and release) which may in turn modulate gene delivery. Only a better understanding of these processes may allow the creation of novel robust engineered systems, potentially opening up a whole new area of biomaterial-guided gene delivery for non-viral systems.
Collapse
Affiliation(s)
- Valeria Graceffa
- Cellular Health and Toxicology Research Group (CHAT), Institute of Technology Sligo, Ash Ln, Bellanode, Sligo, Ireland.
- Department of Life Sciences, Institute of Technology Sligo, Ash Ln, Bellanode, Sligo, Ireland.
| |
Collapse
|
13
|
Kim YS, Mikos AG. Emerging strategies in reprogramming and enhancing the fate of mesenchymal stem cells for bone and cartilage tissue engineering. J Control Release 2021; 330:565-574. [DOI: 10.1016/j.jconrel.2020.12.055] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 12/21/2020] [Accepted: 12/29/2020] [Indexed: 02/06/2023]
|
14
|
Yang R, Chen F, Guo J, Zhou D, Luan S. Recent advances in polymeric biomaterials-based gene delivery for cartilage repair. Bioact Mater 2020; 5:990-1003. [PMID: 32671293 PMCID: PMC7338882 DOI: 10.1016/j.bioactmat.2020.06.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 05/29/2020] [Accepted: 06/10/2020] [Indexed: 12/16/2022] Open
Abstract
Untreated articular cartilage damage normally results in osteoarthritis and even disability that affects millions of people. However, both the existing surgical treatment and tissue engineering approaches are unable to regenerate the original structures of articular cartilage durably, and new strategies for integrative cartilage repair are needed. Gene therapy provides local production of therapeutic factors, especially guided by biomaterials can minimize the diffusion and loss of the genes or gene complexes, achieve accurate spatiotemporally release of gene products, thus provideing long-term treatment for cartilage repair. The widespread application of gene therapy requires the development of safe and effective gene delivery vectors and supportive gene-activated matrices. Among them, polymeric biomaterials are particularly attractive due to their tunable physiochemical properties, as well as excellent adaptive performance. This paper reviews the recent advances in polymeric biomaterial-guided gene delivery for cartilage repair, with an emphasis on the important role of polymeric biomaterials in delivery systems.
Collapse
Affiliation(s)
- Ran Yang
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, PR China
- College of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, PR China
| | - Fei Chen
- Center for Materials Synthetic Biology, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
| | - Jinshan Guo
- Department of Histology and Embryology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, PR China
| | - Dongfang Zhou
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, PR China
| | - Shifang Luan
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China
- College of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, PR China
| |
Collapse
|
15
|
He J, Zhang N, Zhu Y, Jin R, Wu F. MSC spheroids-loaded collagen hydrogels simultaneously promote neuronal differentiation and suppress inflammatory reaction through PI3K-Akt signaling pathway. Biomaterials 2020; 265:120448. [PMID: 33068892 DOI: 10.1016/j.biomaterials.2020.120448] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 10/10/2020] [Indexed: 02/06/2023]
Abstract
It is critical for the clinical success to take the anti-inflammatory function into consideration when integrating the neurogenesis into the nerve repair materials. To this aim, we prepared mesenchymal stem cell (MSC) spheroids-loaded collagen (Col) hydrogels with combined superior anti-inflammatory efficacy and neurogenic activity. The size of the MSC spheroids showed a strong modulation effect on both functions, and the MSC spheroids-100 sample exhibited the best neuronal and anti-inflammatory potentials. The observed dual functions were likely based on the elevated intrinsic cell-cell contacts and cell-extracellular matrix interactions from the MSC spheroids. MSC self-assembly as spheroids expedited the secretions of endogenous trophic factors and extracellular matrix (ECM), which was beneficial to drive neural stem cell differentiation into the neuronal lineage. In addition, the formation of the MSC spheroids secreted more amounts and types of cytokines as well as immunomodulatory paracrine factors to suppress LPS-induced inflammatory reaction. LC-MS/MS analysis further demonstrated that MSC spheroids contributed to the activation of neuroactive ligand-receptor interaction, thereby triggering downstream PI3K-Akt signal pathway, which was likely due to the acceleration of ECM-receptor interaction, gap junction and tight junction. Importantly, inhibiting Akt pathway significantly suppressed the neuronal differentiation, indicating that PI3K-Akt signal pathway was critically involved in the Col-MSC spheroid hydrogel mediated neuroprotection and neurogenesis. Such findings not only provided a simple approach for improving MSC-based therapies for neuron-related diseases, but also shed insight on understanding the underlying mechanisms of MSC-mediated neuronal differentiation.
Collapse
Affiliation(s)
- Jing He
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Nihui Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Yue Zhu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Rongrong Jin
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China.
| | - Fang Wu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China.
| |
Collapse
|
16
|
Xiahou Z, She Y, Zhang J, Qin Y, Li G, Zhang L, Fang H, Zhang K, Chen C, Yin J. Designer Hydrogel with Intelligently Switchable Stem-Cell Contact for Incubating Cartilaginous Microtissues. ACS APPLIED MATERIALS & INTERFACES 2020; 12:40163-40175. [PMID: 32799444 DOI: 10.1021/acsami.0c13426] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Stem-cell-derived organoid can resemble in vivo tissue counterpart and mimic at least one function of tissue or organ, possessing great potential for biomedical application. The present study develops a hydrogel with cell-responsive switch to guide spontaneous and sequential proliferation and aggregation of adipose-derived stem cells (ASCs) without inputting artificial stimulus for in vitro constructing cartilaginous microtissues with enhanced retention of cell-matrix and cell-cell interactions. Polylactic acid (PLA) rods are surface-aminolyzed by cystamine, followed by being involved in the amidation of poly(( l-glutamic acid) and adipic acid dihydrazide (ADH) to form a hydrogel. Along with tubular pore formation in hydrogel after dissolution of PLA rods, aminolyzed PLA molecules with disulfide bonds on rod surfaces are covalently transferred to the tubular pore surfaces of poly(l-glutamic acid)/ADH hydrogel. Because PLA attaches cells, while poly(l-glutamic acid)/ADH hydrogel repels cells, ASCs are found to adhere and proliferate on the tubular pore surfaces of hydrogel first and then cleave disulfide bonds by secreting molecules containing thiol, thus inducing desorption of PLA molecules and leading to their spontaneous detachment and aggregation. Associated with chondrogenic induction by TGF-β1 and IGF-1 in vitro for 28 days, the hydrogel as an all-in-one incubator produces well-engineered columnar cartilage microtissues from ASCs, with the glycosaminoglycans (GAGs) and collagen type II (COL II) deposition achieving 64 and 69% of those in chondrocytes pellet, respectively. The cartilage microtissues further matured in vivo for 8 weeks to exhibit extremely similar histological features and biomechanical performance to native hyaline cartilage. The GAGs and COL II content, as well as compressive modulus of the matured tissue show no significant difference with native cartilage. The designer hydrogel may hold a promise for long-term culture of other types of stem cells and organoids.
Collapse
Affiliation(s)
- Zijie Xiahou
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, Shanghai 200444, P. R. China
| | - Yunlang She
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, P. R. China
| | - Jiahui Zhang
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, Shanghai 200444, P. R. China
| | - Yechi Qin
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, Shanghai 200444, P. R. China
| | - Guifei Li
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, Shanghai 200444, P. R. China
| | - Lili Zhang
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, Shanghai 200444, P. R. China
| | - Haowei Fang
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, Shanghai 200444, P. R. China
| | - Kunxi Zhang
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, Shanghai 200444, P. R. China
- Central Laboratory, Shanghai Putuo Peoples Hospital, Tongji University School of Medicine, Shanghai 200060, P. R. China
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, P. R. China
| | - Jingbo Yin
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, Shanghai 200444, P. R. China
| |
Collapse
|
17
|
Zong H, Wang B, Li G, Yan S, Zhang K, Shou Y, Yin J. Biodegradable High-Strength Hydrogels with Injectable Performance Based on Poly(l-Glutamic Acid) and Gellan Gum. ACS Biomater Sci Eng 2020; 6:4702-4713. [DOI: 10.1021/acsbiomaterials.0c00915] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Hongjie Zong
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, No. 99 Shangda Road, Shanghai 200444, P. R. China
| | - Bo Wang
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, No. 99 Shangda Road, Shanghai 200444, P. R. China
| | - Guifei Li
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, No. 99 Shangda Road, Shanghai 200444, P. R. China
| | - Shifeng Yan
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, No. 99 Shangda Road, Shanghai 200444, P. R. China
| | - Kunxi Zhang
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, No. 99 Shangda Road, Shanghai 200444, P. R. China
| | - Yufeng Shou
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, No. 99 Shangda Road, Shanghai 200444, P. R. China
| | - Jingbo Yin
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, No. 99 Shangda Road, Shanghai 200444, P. R. China
| |
Collapse
|
18
|
mRNA as a Tool for Gene Transfection in 3D Cell Culture for Future Regenerative Therapy. MICROMACHINES 2020; 11:mi11040426. [PMID: 32325734 PMCID: PMC7231348 DOI: 10.3390/mi11040426] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 04/14/2020] [Accepted: 04/16/2020] [Indexed: 01/31/2023]
Abstract
A combination of three-dimensional (3D) cell culturing and non-viral gene transfection is promising in improving outcomes of cell transplantation therapy. Herein, gene transfection profiles in 3D cell culture were compared between plasmid DNA (pDNA) and messenger RNA (mRNA) introduction, using mesenchymal stem cell (MSC) 3D spheroids. Green fluorescence protein (GFP) mRNA induced GFP protein expression in 77% of the cells in the spheroids, whereas only 34% of the cells became GFP positive following pDNA introduction. In mechanistic analyses, most of the cells in MSC spheroids were non-dividing, and pDNA failed to induce GFP expression in most of the non-dividing cells. In contrast, both dividing and non-dividing cells became GFP-positive after mRNA introduction, which led to a high overall percentage of GFP-positive cells in the spheroids. Consequently, mRNA encoding an osteogenic factor, runt-related transcription factor 2 (Runx2), allowed in vitro osteogenic differentiation of MSCs in spheroids more efficiently compared to Runx2 pDNA. Conclusively, mRNA exhibits high potential in gene transfection in 3D cell culture, in which the cell division rate is lower than that in monolayer culture, and the combination of mRNA introduction and 3D cell culture is a promising approach to improve outcomes of cell transplantation in future regenerative therapy.
Collapse
|
19
|
Qin Y, Li G, Wang C, Zhang D, Zhang L, Fang H, Yan S, Zhang K, Yin J. Biomimetic Bilayer Scaffold as an Incubator to Induce Sequential Chondrogenesis and Osteogenesis of Adipose Derived Stem Cells for Construction of Osteochondral Tissue. ACS Biomater Sci Eng 2020; 6:3070-3080. [PMID: 33463252 DOI: 10.1021/acsbiomaterials.0c00200] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Toward osteochondral tissue construction, the present study introduced a bilayer scaffold to induce sequential chondrogenesis and osteogenesis of stem cells in vitro. Two scaffolds that are both based on poly(l-glutamic acid) (PLGA) and chitosan (CS) were combined to form the bilayer scaffold. The cartilage region was the covalently cross-linked PLGA/CS hydrogel with a tubular pore structure, possessing a swollen network to prevent cellular adhesion, while inducing spontaneous cellular aggregate formation. The bone region was the electrostatically cross-linked PLGA-grafted nano hydroxyapatite (nHA-g-PLGA)/CS scaffold, which supported cellular adhesion and spreading. Human adipose derived stem cells (hASCs) were seeded into the cartilage region and observed to aggregate, formimg multicellular spheroids, which subsequently fused to rod-like aggregates with a larger size. At the same time, hASCs in aggregates crossed the interface and entered the bone region, presenting adhesion and spreading. With the induction of bone morphogenetic protein 2 (BMP-2) and insulin-like growth factor 1 (IGF-1) during the first 14 days and BMP-2 alone during the last 14 days, hASCs aggregates in the cartilage region underwent chondrogenesis, expressing an abundant cartilage matrix including glycosaminoglycans (GAGs) and type II collagen (COL II) at 28 days. The chondrogenic induced hASCs migrated in the bone region turned to osteogenesis at 28 days, which was associated with their large spreading area and the switch of the induce factor. Thus, the present bilayer scaffold induced the different distribution of hASCs, resulting in subsequent chondrogenesis and osteogenesis, realizing osteochondral tissue construction in vitro.
Collapse
Affiliation(s)
- Yechi Qin
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, Shanghai 200444, China
| | - Guifei Li
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, Shanghai 200444, China
| | - Chen Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Danqing Zhang
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, Shanghai 200444, China
| | - Lili Zhang
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, Shanghai 200444, China
| | - Haowei Fang
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, Shanghai 200444, China
| | - Shifeng Yan
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, Shanghai 200444, China
| | - Kunxi Zhang
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, Shanghai 200444, China
| | - Jingbo Yin
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, Shanghai 200444, China
| |
Collapse
|
20
|
Kronemberger GS, Matsui RAM, Miranda GDASDCE, Granjeiro JM, Baptista LS. Cartilage and bone tissue engineering using adipose stromal/stem cells spheroids as building blocks. World J Stem Cells 2020; 12:110-122. [PMID: 32184936 PMCID: PMC7062040 DOI: 10.4252/wjsc.v12.i2.110] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 10/19/2019] [Accepted: 01/15/2020] [Indexed: 02/06/2023] Open
Abstract
Scaffold-free techniques in the developmental tissue engineering area are designed to mimic in vivo embryonic processes with the aim of biofabricating, in vitro, tissues with more authentic properties. Cell clusters called spheroids are the basis for scaffold-free tissue engineering. In this review, we explore the use of spheroids from adult mesenchymal stem/stromal cells as a model in the developmental engineering area in order to mimic the developmental stages of cartilage and bone tissues. Spheroids from adult mesenchymal stromal/stem cells lineages recapitulate crucial events in bone and cartilage formation during embryogenesis, and are capable of spontaneously fusing to other spheroids, making them ideal building blocks for bone and cartilage tissue engineering. Here, we discuss data from ours and other labs on the use of adipose stromal/stem cell spheroids in chondrogenesis and osteogenesis in vitro. Overall, recent studies support the notion that spheroids are ideal "building blocks" for tissue engineering by “bottom-up” approaches, which are based on tissue assembly by advanced techniques such as three-dimensional bioprinting. Further studies on the cellular and molecular mechanisms that orchestrate spheroid fusion are now crucial to support continued development of bottom-up tissue engineering approaches such as three-dimensional bioprinting.
Collapse
Affiliation(s)
- Gabriela S Kronemberger
- Laboratory of Tissue Bioengineering, Directory of Metrology Applied to Life Sciences, National Institute of Metrology, Quality and Technology (INMETRO), Duque de Caxias, RJ 25250-020, Brazil
- Post-graduate Program in Translational Biomedicine (Biotrans), Unigranrio, Campus I, Duque de Caxias, RJ 25250-020, Brazil
| | - Renata Akemi Morais Matsui
- Laboratory of Tissue Bioengineering, Directory of Metrology Applied to Life Sciences, National Institute of Metrology, Quality and Technology (INMETRO), Duque de Caxias, RJ 25250-020, Brazil
- Post-graduate Program in Biotechnology, National Institute of Metrology, Quality and Technology (INMETRO), Duque de Caxias, RJ 25250-020, Brazil
| | - Guilherme de Almeida Santos de Castro e Miranda
- Laboratory of Tissue Bioengineering, Directory of Metrology Applied to Life Sciences, National Institute of Metrology, Quality and Technology (INMETRO), Duque de Caxias, RJ 25250-020, Brazil
- Federal University of Rio de Janeiro (UFRJ), Campus Duque de Caxias, Duque de Caxias, RJ 25250-020, Brazil
| | - José Mauro Granjeiro
- Laboratory of Tissue Bioengineering, Directory of Metrology Applied to Life Sciences, National Institute of Metrology, Quality and Technology (INMETRO), Duque de Caxias, RJ 25250-020, Brazil
- Post-graduate Program in Biotechnology, National Institute of Metrology, Quality and Technology (INMETRO), Duque de Caxias, RJ 25250-020, Brazil
- Laboratory of Clinical Research in Odontology, Fluminense Federal University (UFF), Niterói 25255-030 Brazil
| | - Leandra Santos Baptista
- Laboratory of Tissue Bioengineering, Directory of Metrology Applied to Life Sciences, National Institute of Metrology, Quality and Technology (INMETRO), Duque de Caxias, RJ 25250-020, Brazil
- Post-graduate Program in Translational Biomedicine (Biotrans), Unigranrio, Campus I, Duque de Caxias, RJ 25250-020, Brazil
- Post-graduate Program in Biotechnology, National Institute of Metrology, Quality and Technology (INMETRO), Duque de Caxias, RJ 25250-020, Brazil
- Multidisciplinary Center for Biological Research (Numpex-Bio), Federal University of Rio de Janeiro (UFRJ) Campus Duque de Caxias, Duque de Caxias, RJ 25245-390, Brazil
| |
Collapse
|
21
|
Xiao S, Peng Q, Yang Y, Tao Y, Zhou Y, Xu W, Shi X. Preparation of [Amine-Terminated Generation 5 Poly(amidoamine)]-graft-Poly(lactic-co-glycolic acid) Electrospun Nanofibrous Mats for Scaffold-Mediated Gene Transfection. ACS APPLIED BIO MATERIALS 2019; 3:346-357. [PMID: 35019451 DOI: 10.1021/acsabm.9b00848] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Shili Xiao
- School of Textile Science and Engineering, Wuhan Textile University, Wuhan 430200, People’s Republic of China
- State Key Laboratory Cultivation Base for New Textile Materials & Advanced Processing Technology, Wuhan Textile University, 430200 Wuhan, China
| | - Qingyan Peng
- School of Textile Science and Engineering, Wuhan Textile University, Wuhan 430200, People’s Republic of China
| | - Yuhui Yang
- School of Textile Science and Engineering, Wuhan Textile University, Wuhan 430200, People’s Republic of China
| | - Yongzhen Tao
- State Key Laboratory Cultivation Base for New Textile Materials & Advanced Processing Technology, Wuhan Textile University, 430200 Wuhan, China
| | - Yang Zhou
- School of Textile Science and Engineering, Wuhan Textile University, Wuhan 430200, People’s Republic of China
| | - Weilin Xu
- State Key Laboratory Cultivation Base for New Textile Materials & Advanced Processing Technology, Wuhan Textile University, 430200 Wuhan, China
| | - Xiangyang Shi
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, People’s Republic of China
| |
Collapse
|