1
|
Yang X, Mistry M, Chen AD, Chan BP. Tailoring extracellular matrix niches: Impact of glycosaminoglycan content on multiple differentiation of human mesenchymal stem cells. Biomaterials 2025; 318:123130. [PMID: 39893783 DOI: 10.1016/j.biomaterials.2025.123130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 01/12/2025] [Accepted: 01/23/2025] [Indexed: 02/04/2025]
Abstract
Glycosaminoglycan (GAG) represents an important extracellular matrix (ECM), particularly in GAG-rich tissues such as nucleus pulposus and cartilage. The ratio of GAGs/hydroxyproline (HYP) is an indicator of the relative abundance of the space-filling GAG matrix to the fibrous collagen matrix in a particular tissue. Here, we hypothesize that ECM niche with different GAG/HYP ratios will affect the outcomes of multiple differentiation of human mesenchymal stem cells (hMSCs). Specifically, we fabricated collagen-based biomaterials with different GAG/HYP ratios, and differentiate hMSCs in these materials towards osteogenic, chondrogenic and discogenic lineages. In osteogenic differentiation, Collagen without GAG (GAG/HYP ratio 0) showed higher calcium (Ca) and phosphorus (P) deposition and Ca/P ratio, more biomimetic ultrastructure, and better osteogenic phenotypic expression. For chondrogenic differentiation, aminated collagen (aCol-GAG) with intermediate GAG content (GAG/HYP ratio 5.0:1) showed higher GAG deposition, more biomimetic ultrastructure, and better chondrogenic phenotype. In discogenic differentiation, aminated collagen-aminated hyaluronic acid (aHA)-GAG (aCol-aHA-GAG) with the highest GAG content (GAG/HYP ratio 19.8:1), showed intensive GAG deposition, biomimetic ultrastructure, and higher phenotypic marker expression. This study contributes to developing collagen-based biomimetic materials with different GAG/HYP ratios and suggests the use of tissue-specific GAG/HYP ratio as a scaffold design parameter for hMSCs-based musculoskeletal tissue engineering. (198 words).
Collapse
Affiliation(s)
- Xingxing Yang
- Tissue Engineering Lab, School of Biomedical Science, Institute of Tissue Engineering and Regenerative Medicine, Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; Department of Mechanical Engineering, The University of Hong Kong, Hong Kong Special Administrative Region, China; Advanced Biomedical Instrumentation Centre, Hong Kong Special Administrative Region, China
| | - Maitraee Mistry
- Tissue Engineering Lab, School of Biomedical Science, Institute of Tissue Engineering and Regenerative Medicine, Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; Department of Mechanical Engineering, The University of Hong Kong, Hong Kong Special Administrative Region, China; Advanced Biomedical Instrumentation Centre, Hong Kong Special Administrative Region, China
| | - Abigail Dee Chen
- Tissue Engineering Lab, School of Biomedical Science, Institute of Tissue Engineering and Regenerative Medicine, Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; Department of Mechanical Engineering, The University of Hong Kong, Hong Kong Special Administrative Region, China; Advanced Biomedical Instrumentation Centre, Hong Kong Special Administrative Region, China
| | - Barbara Pui Chan
- Tissue Engineering Lab, School of Biomedical Science, Institute of Tissue Engineering and Regenerative Medicine, Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; Department of Mechanical Engineering, The University of Hong Kong, Hong Kong Special Administrative Region, China; Advanced Biomedical Instrumentation Centre, Hong Kong Special Administrative Region, China.
| |
Collapse
|
2
|
Zhang H, Zhou Z, Zhang F, Wan C. Hydrogel-Based 3D Bioprinting Technology for Articular Cartilage Regenerative Engineering. Gels 2024; 10:430. [PMID: 39057453 PMCID: PMC11276275 DOI: 10.3390/gels10070430] [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: 05/13/2024] [Revised: 06/09/2024] [Accepted: 06/21/2024] [Indexed: 07/28/2024] Open
Abstract
Articular cartilage is an avascular tissue with very limited capacity of self-regeneration. Trauma or injury-related defects, inflammation, or aging in articular cartilage can induce progressive degenerative joint diseases such as osteoarthritis. There are significant clinical demands for the development of effective therapeutic approaches to promote articular cartilage repair or regeneration. The current treatment modalities used for the repair of cartilage lesions mainly include cell-based therapy, small molecules, surgical approaches, and tissue engineering. However, these approaches remain unsatisfactory. With the advent of three-dimensional (3D) bioprinting technology, tissue engineering provides an opportunity to repair articular cartilage defects or degeneration through the construction of organized, living structures composed of biomaterials, chondrogenic cells, and bioactive factors. The bioprinted cartilage-like structures can mimic native articular cartilage, as opposed to traditional approaches, by allowing excellent control of chondrogenic cell distribution and the modulation of biomechanical and biochemical properties with high precision. This review focuses on various hydrogels, including natural and synthetic hydrogels, and their current developments as bioinks in 3D bioprinting for cartilage tissue engineering. In addition, the challenges and prospects of these hydrogels in cartilage tissue engineering applications are also discussed.
Collapse
Affiliation(s)
- Hongji Zhang
- Key Laboratory of Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; (H.Z.); (Z.Z.); (F.Z.)
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Hong Kong SAR, China
- Key Laboratory of Regenerative Medicine (Shenzhen Base), Ministry of Education, School of Biomedical Sciences Core Laboratory, Institute of Stem Cell, Genomics and Translational Research, Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen 518057, China
| | - Zheyuan Zhou
- Key Laboratory of Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; (H.Z.); (Z.Z.); (F.Z.)
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Hong Kong SAR, China
- Key Laboratory of Regenerative Medicine (Shenzhen Base), Ministry of Education, School of Biomedical Sciences Core Laboratory, Institute of Stem Cell, Genomics and Translational Research, Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen 518057, China
| | - Fengjie Zhang
- Key Laboratory of Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; (H.Z.); (Z.Z.); (F.Z.)
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Hong Kong SAR, China
- Key Laboratory of Regenerative Medicine (Shenzhen Base), Ministry of Education, School of Biomedical Sciences Core Laboratory, Institute of Stem Cell, Genomics and Translational Research, Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen 518057, China
| | - Chao Wan
- Key Laboratory of Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; (H.Z.); (Z.Z.); (F.Z.)
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Hong Kong SAR, China
- Key Laboratory of Regenerative Medicine (Shenzhen Base), Ministry of Education, School of Biomedical Sciences Core Laboratory, Institute of Stem Cell, Genomics and Translational Research, Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen 518057, China
| |
Collapse
|
3
|
Chen X, Huang S, Niu Y, Luo M, Liu H, Jiao Y, Huang J. Transplantation of Gelatin Microspheres Loaded with Wharton's Jelly Derived Mesenchymal Stem Cells Facilitates Cartilage Repair in Mice. Tissue Eng Regen Med 2024; 21:171-183. [PMID: 37688747 PMCID: PMC10764672 DOI: 10.1007/s13770-023-00574-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 06/20/2023] [Accepted: 07/05/2023] [Indexed: 09/11/2023] Open
Abstract
BACKGROUND Knee osteoarthritis (KOA) is a prevalent chronic joint disease caused by various factors. Mesenchymal stem cells (MSCs) therapy is an increasingly promising therapeutic option for osteoarthritis. However, the chronic inflammation of knee joint can severely impede the therapeutic effects of transplanted cells. Gelatin microspheres (GMs) are degradable biomaterial that have various porosities for cell adhesion and cell-cell interaction. Excellent elasticity and deformability of GMs make it an excellent injectable vehicle for cell delivery. METHODS We created Wharton's jelly derived mesenchymal stem cells (WJMSCs)-GMs complexes and assessed the effects of GMs on cell activity, proliferation and chondrogenesis. Then, WJMSCs loaded in GMs were transplanted in the joint of osteoarthritis mice. After four weeks, joint tissue was collected for histological analysis. Overexpressing-luciferase WJMSCs were performed to explore cell retention in mice. RESULTS In vitro experiments demonstrated that WJMSCs loaded with GMs maintained cell viability and proliferative potential. Moreover, GMs enhanced the chondrogenesis differentiation of WJMSCs while alleviated cell hypertrophy. In KOA mice model, transplantation of WJMSCs-GMs complexes promoted cartilage regeneration and cartilage matrix formation, contributing to the treatment of KOA. Compared with other groups, in WJMSCs+GMs group, there were fewer cartilage defects and with a more integrated tibia structure. Tracking results of stable-overexpressing luciferase WJMSCs demonstrated that GMs significantly extended the retention time of WJMSCs in knee joint cavity. CONCLUSION Our results indicated that GMs facilitate WJMSCs mediated knee osteoarthritis healing in mice by promoting cartilage regeneration and prolonging cell retention. It might potentially provide an optimal strategy for the biomaterial-stem cell based therapy for knee osteoarthritis.
Collapse
Affiliation(s)
- Xiaolin Chen
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Sunxing Huang
- Key Laboratory of Reproductive Medicine of Guangdong Province, The First Affliated Hospital and School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Yongxia Niu
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Mingxun Luo
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Haiying Liu
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Yiren Jiao
- Joint Centre of Translational Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China.
| | - Junjiu Huang
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China.
- Key Laboratory of Reproductive Medicine of Guangdong Province, The First Affliated Hospital and School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China.
| |
Collapse
|
4
|
Gao Y, Dai W, Li S, Zhao X, Wang J, Fu W, Guo L, Fan Y, Zhang X. Components and physical properties of hydrogels modulate inflammatory response and cartilage repair. J Mater Chem B 2023; 11:10029-10042. [PMID: 37850311 DOI: 10.1039/d3tb01917a] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2023]
Abstract
Collagen and hyaluronic acid are commonly applied in cartilage tissue engineering, yet there has been limited investigation into their inflammatory response, a crucial factor in articular cartilage repair. This study aimed to evaluate the impact of components and physical properties of hydrogels on inflammatory response and cartilage repair. Three kinds of hydrogels with comparable storage moduli at low frequencies were designed and fabricated, namely, methacrylic anhydride-modified hyaluronic acid hydrogel (HAMA), methacrylic anhydride-modified type I collagen hydrogel (CMA) and unmodified type I collagen hydrogel (Col). HAMA hydrogel was unfavorable for adhesion and spreading of BMSCs. Furthermore, HAMA hydrogel stimulated rapid migration and pro-inflammatory M1 polarization of macrophages, leading to persistent and intense inflammation, which was unfavorable for cartilage repair. CMA and Col hydrogels possessed the same component and facilitated the adhesion, spreading and proliferation of BMSCs. Compared with CMA hydrogel, Col hydrogel induced rapid migration and moderate M1 polarization of macrophages at the early stage of injury, which was mainly influenced by its fast dissolution rate, small pore size fiber network structure and rapid stress relaxation. In addition, the phenotype of macrophages timely transformed into anti-inflammatory M2 due to the properties of the collagen component, which shortened the duration of inflammation and enhanced cartilage repair. The results indicated that moderate macrophage activation adjusted by hydrogel components and physical properties was critical in modulating inflammation and cartilage regeneration.
Collapse
Affiliation(s)
- Yongli Gao
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, China.
- School of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, China
| | - Wenling Dai
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, China.
- School of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, China
| | - Shikui Li
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, China.
- School of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, China
| | - Xingchen Zhao
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, China.
- School of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, China
| | - Jing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, China.
- School of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, China
| | - Weili Fu
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan 610064, China
| | - Likun Guo
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, China.
- School of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, China
| | - Yujiang Fan
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, China.
- School of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, China.
- School of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan 610064, China
| |
Collapse
|
5
|
Fan S, Sun X, Su C, Xue Y, Song X, Deng R. Macrophages-bone marrow mesenchymal stem cells crosstalk in bone healing. Front Cell Dev Biol 2023; 11:1193765. [PMID: 37427382 PMCID: PMC10327485 DOI: 10.3389/fcell.2023.1193765] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 06/14/2023] [Indexed: 07/11/2023] Open
Abstract
Bone healing is associated with many orthopedic conditions, including fractures and osteonecrosis, arthritis, metabolic bone disease, tumors and periprosthetic particle-associated osteolysis. How to effectively promote bone healing has become a keen topic for researchers. The role of macrophages and bone marrow mesenchymal stem cells (BMSCs) in bone healing has gradually come to light with the development of the concept of osteoimmunity. Their interaction regulates the balance between inflammation and regeneration, and when the inflammatory response is over-excited, attenuated, or disturbed, it results in the failure of bone healing. Therefore, an in-depth understanding of the function of macrophages and bone marrow mesenchymal stem cells in bone regeneration and the relationship between the two could provide new directions to promote bone healing. This paper reviews the role of macrophages and bone marrow mesenchymal stem cells in bone healing and the mechanism and significance of their interaction. Several new therapeutic ideas for regulating the inflammatory response in bone healing by targeting macrophages and bone marrow mesenchymal stem cells crosstalk are also discussed.
Collapse
Affiliation(s)
- Siyu Fan
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Xin Sun
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Chuanchao Su
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Yiwen Xue
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Xiao Song
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Runzhi Deng
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| |
Collapse
|
6
|
Yang KC, Yang YT, Wu CC, Hsiao JK, Huang CY, Chen IH, Wang CC. Bioinspired collagen-gelatin-hyaluronic acid-chondroitin sulfate tetra-copolymer scaffold biomimicking native cartilage extracellular matrix facilitates chondrogenesis of human synovium-derived stem cells. Int J Biol Macromol 2023; 240:124400. [PMID: 37044324 DOI: 10.1016/j.ijbiomac.2023.124400] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 03/15/2023] [Accepted: 04/06/2023] [Indexed: 04/14/2023]
Abstract
The microenvironment plays a crucial role in stem cell differentiation, and a scaffold that mimics native cartilaginous extracellular components can promote chondrogenesis. In this study, a collagen-gelatin-hyaluronic acid-chondroitin sulfate tetra-copolymer scaffold with composition and architecture similar to those of hyaline cartilage was fabricated using a microfluidic technique and compared with a pure gelatin scaffold. The newly designed biomimetic scaffold had a swelling ratio of 1278 % ± 270 %, a porosity of 77.68 % ± 11.70 %, a compressive strength of 1005 ± 174 KPa, and showed a good resilience against compression force. Synovium-derived stem cells (SDSCs) seeded into the tetra-copolymer scaffold attached to the scaffold firmly and exhibited good mitochondrial activity, high cell survival with a pronounced glycosaminoglycan production. SDSCs cultured on the tetra-copolymer scaffold with chondrogenic induction exhibited upregulated mRNA expression of COL2A1, ChM-1, Nrf2, TGF-β1, and BMP-7. Ex vivo study revealed that the SDSC-tetra-copolymer scaffold regenerated cartilage-like tissue in SCID mice with abundant type II collagen and S-100 production. BMP7 and COL2A1 expression in the tetra-copolymer scaffold group was much higher than that in the gelatin scaffold group ex vivo. The tetra-copolymer scaffold thus exhibits strong chondrogenic capability and will facilitate cartilage tissue engineering.
Collapse
Affiliation(s)
- Kai-Chiang Yang
- Department of Orthopedic Surgery, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231016, Taiwan; School of Dental Technology, College of Oral Medicine, Taipei Medical University, Taipei 110301, Taiwan; Department of Orthopedics, En Chu Kong Hospital, New Taipei City 237011, Taiwan
| | - Ya-Ting Yang
- Department of Orthopedic Surgery, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231016, Taiwan
| | - Chang-Chin Wu
- Department of Orthopedics, En Chu Kong Hospital, New Taipei City 237011, Taiwan; Departments of Biomedical Engineering, Yuanpei University of Medical Technology, Hsinchu City 300102, Taiwan
| | - Jong-Kai Hsiao
- Department of Medical Imaging, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231016, Taiwan
| | - Chien-Yuan Huang
- Department of Orthopedic Surgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung City 427213, Taiwan
| | - Ing-Ho Chen
- Department of Orthopedic Surgery, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231016, Taiwan; Department of Orthopedic Surgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970473, Taiwan; Department of Orthopedics, School of Medicine, Tzu Chi University, Hualien 970374, Taiwan
| | - Chen-Chie Wang
- Department of Orthopedic Surgery, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231016, Taiwan; Department of Orthopedics, School of Medicine, Tzu Chi University, Hualien 970374, Taiwan.
| |
Collapse
|
7
|
Yang XX, Yip CH, Zhao S, Ho YP, Chan BP. A bio-inspired nano-material recapitulating the composition, ultra-structure, and function of the glycosaminoglycan-rich extracellular matrix of nucleus pulposus. Biomaterials 2023; 293:121991. [PMID: 36586145 DOI: 10.1016/j.biomaterials.2022.121991] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 12/22/2022] [Accepted: 12/24/2022] [Indexed: 12/28/2022]
Abstract
The nucleus pulposus (NP) of intervertebral disc represents a soft gel consisting of glycosaminoglycans (GAGs)-rich extracellular matrix (ECM). Significant loss of GAGs and normal functions are the most prevalent changes in degenerated disc. Attempts targeted to incorporate GAGs into collagen fibrous matrices have been made but the efficiency is very low, and the resulting structures showed no similarity with native NP. Inspired by the characteristic composition and structures of the ECM of native NP, here, we hypothesize that by chemically modifying the collagen (Col) and hyaluronic acid (HA) and co-precipitating with GAGs, a bio-inspired nano-material recapitulating the composition, ultra-structure and function of the GAG-rich ECM will be fabricated. Compositionally, the bio-inspired nano-material namely Aminated Collagen-Aminated Hyaluronic Acid-GAG (aCol-aHA-GAG) shows a record high GAG/hydroxyproline ratio up to 39.1:1 in a controllable manner, out-performing that of the native NP. Ultra-structurally, the nano-material recapitulates the characteristic 'nano-beads' (25 nm) and 'bottle-brushes' (133 nm) features as those found in native NP. Functionally, the nano-material supports the viability and maintains the morphological and phenotypic markers of bovine NP cells, and shows comparable mechanical properties of native NP. This work contributes to the development of a compositionally, structurally, and functionally biomimetic nano-material for NP tissue engineering.
Collapse
Affiliation(s)
- Xing-Xing Yang
- Tissue Engineering Laboratory, Department of Mechanical Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong Special Administrative Region
| | - Chi-Hung Yip
- Tissue Engineering Laboratory, Department of Mechanical Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong Special Administrative Region
| | - Shirui Zhao
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Yi-Ping Ho
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Barbara Pui Chan
- Tissue Engineering Laboratory, Department of Mechanical Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong Special Administrative Region.
| |
Collapse
|
8
|
Shen Y, Ning J, Zhao L, Liu W, Wang T, Yu J, Wang Y. Matrix remodeling associated 7 proteins promote cutaneous wound healing through vimentin in coordinating fibroblast functions. Inflamm Regen 2023; 43:5. [PMID: 36647132 PMCID: PMC9841631 DOI: 10.1186/s41232-023-00256-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 01/05/2023] [Indexed: 01/18/2023] Open
Abstract
Wound healing depends largely on the remodeling of the extracellular matrix around and reorganization of tissue-resident cells. Matrix remodeling associated 7 (MXRA7) is a member of the matrix remodeling-associated gene family and is involved in matrix remodeling-associated processes, such as inflammatory neovascularization, liver injury, and autoimmune skin disease. To investigate whether and how MXRA7 participate in cutaneous wound healing, an ear-punching model was utilized in wild-type (WT) and MXRA7-deficient mice, and the dermal fibroblasts from these mice were further studied in vitro. Results showed that the MXRA7 deficiency impaired the wound healing process in mice. Quantitative PCR indicated that lack of MXRA7 impaired the expression of several extracellular matrix genes (e.g., MMP-2) and inhibited signaling pathways (e.g., STAT3) in healing ear tissues. In in vitro culture system, migration, contraction, or proliferation of fibroblasts was impaired upon MXRA7 deficiency. Pull-down and mass spectrum assay revealed that vimentin was among the proteins that bound MXRA7 proteins in cells, and further investigations indicate MXRA7 was an autocrine factor in fibroblasts that involved vimentin in certain ways, such as JNK and STAT3/STAT5 signaling pathways in our study. In conclusion, MXRA7 proteins promote wound healing through vimentin in coordinating fibroblast functions.
Collapse
Affiliation(s)
- Ying Shen
- grid.263761.70000 0001 0198 0694National Clinical Research Center for Hematologic Disease, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou Medical College, Soochow University, Suzhou, 215006 China
| | - Jinling Ning
- grid.263761.70000 0001 0198 0694National Clinical Research Center for Hematologic Disease, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou Medical College, Soochow University, Suzhou, 215006 China
| | - Lu Zhao
- grid.263761.70000 0001 0198 0694National Clinical Research Center for Hematologic Disease, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou Medical College, Soochow University, Suzhou, 215006 China
| | - Wei Liu
- grid.429222.d0000 0004 1798 0228Department of Pathology, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006 China
| | - Ting Wang
- grid.263761.70000 0001 0198 0694National Clinical Research Center for Hematologic Disease, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou Medical College, Soochow University, Suzhou, 215006 China
| | - Jie Yu
- grid.429222.d0000 0004 1798 0228Department of Pathology, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006 China
| | - Yiqiang Wang
- grid.263761.70000 0001 0198 0694National Clinical Research Center for Hematologic Disease, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou Medical College, Soochow University, Suzhou, 215006 China ,grid.12955.3a0000 0001 2264 7233Eye Institute of Xiamen University, Xiamen University, Xiamen, 361104 China ,grid.440701.60000 0004 1765 4000Wisdom Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool University, Suzhou, 215123 China
| |
Collapse
|
9
|
Micropattern Silk Fibroin Film Facilitates Tendon Repair In Vivo and Promotes Tenogenic Differentiation of Tendon Stem/Progenitor Cells through the α2 β1/FAK/PI3K/AKT Signaling Pathway In Vitro. Stem Cells Int 2023; 2023:2915826. [PMID: 36684388 PMCID: PMC9859702 DOI: 10.1155/2023/2915826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 11/23/2022] [Accepted: 12/08/2022] [Indexed: 01/15/2023] Open
Abstract
Background Tendon injuries are common clinical disorders. Due to the limited regeneration ability of tendons, tissue engineering technology is often used as an adjuvant treatment. This study explored the molecular pathways underlying micropattern SF film-regulated TSPC propensity and their repairing effects to highlight the application value of micropattern SF films. Methods First, we characterized the physical properties of the micropattern SF films and explored their repairing effects on the injured tendons in vivo. Then, we seeded TSPCs on SF films in vitro and determined the micropattern SF film-induced gene expression and activation of signaling pathways in TSPCs through high-throughput RNA sequencing and proteomics assays. Results The results of in vivo studies suggested that micropattern SF films can promote remodeling of the injured tendon. In addition, immunohistochemistry (IHC) results showed that tendon marker genes were significantly increased in the micropattern SF film repair group. Transcriptomic and proteomic analyses demonstrated that micropattern SF film-induced genes and proteins in TSPCs were mainly enriched in the focal adhesion kinase (FAK)/actin and phosphoinositide 3-kinase (PI3K)/AKT pathways. Western blot analysis showed that the expression of integrins α2β1, tenascin-C (TNC), and tenomodulin (TNMD) and the phosphorylation of AKT were significantly increased in the micropattern SF film group, which could be abrogated by applying PI3K/AKT inhibitors. Conclusion Micropattern SF films modified by water annealing can promote remodeling of the injured tendon in vivo and regulate the tendon differentiation of TSPCs through the α2β1/FAK/PI3K/AKT signaling pathway in vitro. Therefore, they have great medical value in tendon repair.
Collapse
|
10
|
Strecanska M, Danisovic L, Ziaran S, Cehakova M. The Role of Extracellular Matrix and Hydrogels in Mesenchymal Stem Cell Chondrogenesis and Cartilage Regeneration. LIFE (BASEL, SWITZERLAND) 2022; 12:life12122066. [PMID: 36556431 PMCID: PMC9784885 DOI: 10.3390/life12122066] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/02/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022]
Abstract
Diseases associated with articular cartilage disintegration or loss are still therapeutically challenging. The traditional treatment approaches only alleviate the symptoms while potentially causing serious side effects. The limited self-renewal potential of articular cartilage provides opportunities for advanced therapies involving mesenchymal stem cells (MSCs) that are characterized by a remarkable regenerative capacity. The chondrogenic potential of MSCs is known to be regulated by the local environment, including soluble factors and the less discussed extracellular matrix (ECM) components. This review summarizes the process of chondrogenesis, and also the biological properties of the ECM mediated by mechanotransduction as well as canonical and non-canonical signaling. Our focus is also on the influence of the ECM's physical parameters, molecular composition, and chondrogenic factor affinity on the adhesion, survival, and chondrogenic differentiation of MSCs. These basic biological insights are crucial for a more precise fabrication of ECM-mimicking hydrogels to improve cartilage tissue reconstruction. Lastly, we provide an overview of hydrogel classification and characterization. We also include the results from preclinical models combining MSCs with hydrogels for the treatment of cartilage defects, to support clinical application of this construct. Overall, it is believed that the proper combination of MSCs, hydrogels, and chondrogenic factors can lead to complex cartilage regeneration.
Collapse
Affiliation(s)
- Magdalena Strecanska
- National Institute of Rheumatic Diseases, Nabrezie I. Krasku 4, 921 12 Piestany, Slovakia
- Institute of Medical Biology, Genetics, and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Lubos Danisovic
- National Institute of Rheumatic Diseases, Nabrezie I. Krasku 4, 921 12 Piestany, Slovakia
- Institute of Medical Biology, Genetics, and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Stanislav Ziaran
- National Institute of Rheumatic Diseases, Nabrezie I. Krasku 4, 921 12 Piestany, Slovakia
- Department of Urology, Faculty of Medicine, Comenius University, Limbova 5, 833 05 Bratislava, Slovakia
| | - Michaela Cehakova
- National Institute of Rheumatic Diseases, Nabrezie I. Krasku 4, 921 12 Piestany, Slovakia
- Institute of Medical Biology, Genetics, and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
- Correspondence: ; Tel.: +421-2-5935-7215
| |
Collapse
|
11
|
Zhao M, Gao X, Wei J, Tu C, Zheng H, Jing K, Chu J, Ye W, Groth T. Chondrogenic differentiation of mesenchymal stem cells through cartilage matrix-inspired surface coatings. Front Bioeng Biotechnol 2022; 10:991855. [PMID: 36246378 PMCID: PMC9557131 DOI: 10.3389/fbioe.2022.991855] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 09/14/2022] [Indexed: 11/13/2022] Open
Abstract
The stem cell niche comprises soluble molecules and extracellular matrix components which provide chemical and mechanical cues that determine the differentiation of stem cells. Here, the effect of polyelectrolyte multilayer (PEM) composition and terminal layer fabricated with layer-by-layer technique (LBL) pairing either hyaluronan [in its native (nHA) and oxidized form (oHA)] or chondroitin sulfate (CS) with type I collagen (Col I) is investigated on chondrogenic differentiation of human umbilical mesenchymal stem cells (hUC-MSCs). Physical studies performed to investigate the establishment and structure of the surface coatings show that PEM composed of HA and Col I show a dominance of nHA or oHA with considerably lesser organization of Col I fibrils. In contrast, distinguished fibrilized Col I is found in nCS-containing PEM. Generally, Col I-terminated PEM promote the adhesion, migration, and growth of hUC-MSCs more than GAG-terminated surfaces due to the presence of fibrillar Col I but show a lower degree of differentiation towards the chondrogenic lineage. Notably, the Col I/nHA PEM not only supports adhesion and growth of hUC-MSCs but also significantly promotes cartilage-associated gene and protein expression as found by histochemical and molecular biology studies, which is not seen on the Col I/oHA PEM. This is related to ligation of HA to the cell receptor CD44 followed by activation of ERK/Sox9 and noncanonical TGF-β signaling-p38 pathways that depends on the molecular weight of HA as found by immune histochemical and western blotting. Hence, surface coatings on scaffolds and other implants by PEM composed of nHA and Col I may be useful for programming MSC towards cartilage regeneration.
Collapse
Affiliation(s)
- Mingyan Zhao
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- *Correspondence: Mingyan Zhao, ; Thomas Groth,
| | - Xiang Gao
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Jinsong Wei
- Department of Spinal Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Chenlin Tu
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Department of Spinal Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Hong Zheng
- Department of Spinal Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Kaipeng Jing
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Jiaqi Chu
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Wei Ye
- Department of Obstetrics and Gynecology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Thomas Groth
- Department Biomedical Materials, Institute of Pharmacy, Martin Luther University Halle Wittenberg, Halle (Saale), Germany
- *Correspondence: Mingyan Zhao, ; Thomas Groth,
| |
Collapse
|
12
|
Hou M, Tian B, Bai B, Ci Z, Liu Y, Zhang Y, Zhou G, Cao Y. Dominant role of in situ native cartilage niche for determining the cartilage type regenerated by BMSCs. Bioact Mater 2022; 13:149-160. [PMID: 35224298 PMCID: PMC8843973 DOI: 10.1016/j.bioactmat.2021.11.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 11/03/2021] [Accepted: 11/04/2021] [Indexed: 12/27/2022] Open
Affiliation(s)
- Mengjie Hou
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, PR China
- National Tissue Engineering Center of China, Shanghai, PR China
| | - Baoxing Tian
- Department of Breast Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, PR China
| | - Baoshuai Bai
- National Tissue Engineering Center of China, Shanghai, PR China
- Research Institute of Plastic Surgery, Wei Fang Medical College, Weifang, PR China
| | - Zheng Ci
- National Tissue Engineering Center of China, Shanghai, PR China
- Research Institute of Plastic Surgery, Wei Fang Medical College, Weifang, PR China
| | - Yu Liu
- National Tissue Engineering Center of China, Shanghai, PR China
- Research Institute of Plastic Surgery, Wei Fang Medical College, Weifang, PR China
| | - Yixin Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, PR China
| | - Guangdong Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, PR China
- National Tissue Engineering Center of China, Shanghai, PR China
- Research Institute of Plastic Surgery, Wei Fang Medical College, Weifang, PR China
- Corresponding author. Shanghai Key Lab of Tissue Engineering, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, PR China.
| | - Yilin Cao
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Tissue Engineering, Shanghai, PR China
- National Tissue Engineering Center of China, Shanghai, PR China
- Corresponding author. Shanghai Key Lab of Tissue Engineering, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639 Zhi Zao Ju Road, Shanghai, 200011, PR China.
| |
Collapse
|
13
|
Wang H, Hu B, Li H, Feng G, Pan S, Chen Z, Li B, Song J. Biomimetic Mineralized Hydroxyapatite Nanofiber-Incorporated Methacrylated Gelatin Hydrogel with Improved Mechanical and Osteoinductive Performances for Bone Regeneration. Int J Nanomedicine 2022; 17:1511-1529. [PMID: 35388269 PMCID: PMC8978691 DOI: 10.2147/ijn.s354127] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 03/15/2022] [Indexed: 12/16/2022] Open
Abstract
Purpose Methacrylic anhydride-modified gelatin (GelMA) hydrogels exhibit many beneficial biological features and are widely studied for bone tissue regeneration. However, deficiencies in the mechanical strength, osteogenic factors and mineral ions limit their application in bone defect regeneration. Incorporation of inorganic fillers into GelMA to improve its mechanical properties and bone regenerative ability has been one of the research hotspots. Methods In this work, hydroxyapatite nanofibers (HANFs) were prepared and mineralized in a simulated body fluid to make their components and structure more similar to those of natural bone apatite, and then different amounts of mineralized HANFs (m-HANFs) were incorporated into the GelMA hydrogel to form m-HANFs/GelMA composite hydrogels. The physicochemical properties, biocompatibility and bone regenerative ability of m-HANFs/GelMA were determined in vitro and in vivo. Results The results indicated that m-HANFs with high aspect ratio presented rough and porous surfaces coated with bone-like apatite crystals. The incorporation of biomimetic m-HANFs improved the biocompatibility, mechanical, swelling, degradation and bone regenerative performances of GelMA. However, the improvement in the performance of the composite hydrogel did not continuously increase as the amount of added m-HANFs increased, and the 15m-HANFs/GelMA group exhibited the best swelling and degradation performances and the best bone repair effect in vivo among all the groups. Conclusion The biomimetic m-HANFs/GelMA composite hydrogel can provide a novel option for bone tissue engineering in the future; however, it needs further investigations to optimize the proportions of m-HANFs and GelMA for improving the bone repair effect.
Collapse
Affiliation(s)
- He Wang
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, People’s Republic of China
| | - Bo Hu
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, People’s Republic of China
| | - Hong Li
- Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection Technology, Chongqing University of Science and Technology, Chongqing, People’s Republic of China
| | - Ge Feng
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, People’s Republic of China
| | - Shengyuan Pan
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, People’s Republic of China
| | - Ziqi Chen
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, People’s Republic of China
| | - Bo Li
- Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection Technology, Chongqing University of Science and Technology, Chongqing, People’s Republic of China
- Correspondence: Bo Li, Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection Technology, Chongqing University of Science and Technology, Chongqing, 401331, People’s Republic of China, Tel +86-23-8886-0026, Fax +86-23-8886-0222, Email
| | - Jinlin Song
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, People’s Republic of China
- Jinlin Song, College of Stomatology, Chongqing Medical University, 426# Songshibei Road, Yubei District, Chongqing, 401147, People’s Republic of China, Tel +86-23-8886-0026, Fax +86-23-8886-0222, Email
| |
Collapse
|
14
|
Heparin Immobilization of Tissue Engineered Xenogeneic Small Diameter Arterial Scaffold Improve Endothelialization. Tissue Eng Regen Med 2022; 19:505-523. [PMID: 35092597 PMCID: PMC9130405 DOI: 10.1007/s13770-021-00411-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/24/2021] [Accepted: 11/01/2021] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Autologous vessels graft (Inner diameter < 6 mm) harvesting always challenged during bypass grafting surgery and its complication shows poor outcome. Tissue engineered vascular graft allow to generate biological graft without any immunogenic complication. The approach presented in this study is to induce graft remodeling through heparin coating in luminal surface of small diameter (Inner diameter < 1 mm) decellularized arterial graft. METHODS Decellularization of graft was done using SDS, combination of 0.5% sodium dodecyl sulfate and 0.5% sodium deoxycholate and only sodium deoxycholate. Decellularization was confirmed on basis of histology, and DAPI. Characterization of extracellular matrix was analyzed using histology and scanning electron microscopy. Surface modification of decellularized vascular graft was done with heparin coating. Heparin immobilization was evaluated by toluidine blue stain. Heparin-coated graft was transplanted end to end anastomosis in femoral artery in rat. RESULTS Combination of 0.5% sodium dodecyl sulfate and 0.5% Sodium deoxycholate showed complete removal of xenogeneic cells. The heparin coating on luminal surface showed anti-thrombogenicity and endothelialization. Mechanical testing revealed no significant differences in strain characteristics and modulus between native tissues, decellularized scaffolds and transplanted scaffold. Collectively, this study proposed a heparin-immobilized ECM coating to surface modification offering functionalize biomaterials for developing small-diameter vascular grafts. CONCLUSION We conclude that xenogeneic decellularized arterial scaffold with heparin surface modification can be fabricated and successfully transplanted small diameter (inner diameter < 1 mm) decellularized arterial graft.
Collapse
|
15
|
Klarmann GJ, Gaston J, Ho VB. A review of strategies for development of tissue engineered meniscal implants. BIOMATERIALS AND BIOSYSTEMS 2021; 4:100026. [PMID: 36824574 PMCID: PMC9934480 DOI: 10.1016/j.bbiosy.2021.100026] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 08/17/2021] [Accepted: 08/25/2021] [Indexed: 12/09/2022] Open
Abstract
The meniscus is a key stabilizing tissue of the knee that facilitates proper tracking and movement of the knee joint and absorbs stresses related to physical activity. This review article describes the biology, structure, and functions of the human knee meniscus, common tears and repair approaches, and current research and development approaches using modern methods to fabricate a scaffold or tissue engineered meniscal replacement. Meniscal tears are quite common, often resulting from sports or physical training, though injury can result without specific contact during normal physical activity such as bending or squatting. Meniscal injuries often require surgical intervention to repair, restore basic functionality and relieve pain, and severe damage may warrant reconstruction using allograft transplants or commercial implant devices. Ongoing research is attempting to develop alternative scaffold and tissue engineered devices using modern fabrication techniques including three-dimensional (3D) printing which can fabricate a patient-specific meniscus replacement. An ideal meniscal substitute should have mechanical properties that are close to that of natural human meniscus, and also be easily adapted for surgical procedures and fixation. A better understanding of the organization and structure of the meniscus as well as its potential points of failure will lead to improved design approaches to generate a suitable and functional replacement.
Collapse
Affiliation(s)
- George J. Klarmann
- 4D Bio³ Center, Department of Radiology and Radiological Sciences, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Rd., Bethesda, MD 20814, USA,The Geneva Foundation, 917 Pacific Ave., Tacoma, WA 98402, USA,Corresponding author at: USU-4D Bio³ Center, 9410 Key West Ave., Rockville, MD 20850, USA.
| | - Joel Gaston
- 4D Bio³ Center, Department of Radiology and Radiological Sciences, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Rd., Bethesda, MD 20814, USA,The Geneva Foundation, 917 Pacific Ave., Tacoma, WA 98402, USA
| | - Vincent B. Ho
- 4D Bio³ Center, Department of Radiology and Radiological Sciences, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Rd., Bethesda, MD 20814, USA
| |
Collapse
|
16
|
Pozzi S, Scomparin A, Israeli Dangoor S, Rodriguez Ajamil D, Ofek P, Neufeld L, Krivitsky A, Vaskovich-Koubi D, Kleiner R, Dey P, Koshrovski-Michael S, Reisman N, Satchi-Fainaro R. Meet me halfway: Are in vitro 3D cancer models on the way to replace in vivo models for nanomedicine development? Adv Drug Deliv Rev 2021; 175:113760. [PMID: 33838208 DOI: 10.1016/j.addr.2021.04.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 03/24/2021] [Accepted: 04/01/2021] [Indexed: 12/12/2022]
Abstract
The complexity and diversity of the biochemical processes that occur during tumorigenesis and metastasis are frequently over-simplified in the traditional in vitro cell cultures. Two-dimensional cultures limit researchers' experimental observations and frequently give rise to misleading and contradictory results. Therefore, in order to overcome the limitations of in vitro studies and bridge the translational gap to in vivo applications, 3D models of cancer were developed in the last decades. The three dimensions of the tumor, including its cellular and extracellular microenvironment, are recreated by combining co-cultures of cancer and stromal cells in 3D hydrogel-based growth factors-inclusive scaffolds. More complex 3D cultures, containing functional blood vasculature, can integrate in the system external stimuli (e.g. oxygen and nutrient deprivation, cytokines, growth factors) along with drugs, or other therapeutic compounds. In this scenario, cell signaling pathways, metastatic cascade steps, cell differentiation and self-renewal, tumor-microenvironment interactions, and precision and personalized medicine, are among the wide range of biological applications that can be studied. Here, we discuss a broad variety of strategies exploited by scientists to create in vitro 3D cancer models that resemble as much as possible the biology and patho-physiology of in vivo tumors and predict faithfully the treatment outcome.
Collapse
Affiliation(s)
- Sabina Pozzi
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Anna Scomparin
- Department of Drug Science and Technology, University of Turin, Via P. Giuria 9, 10125 Turin, Italy
| | - Sahar Israeli Dangoor
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Daniel Rodriguez Ajamil
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Paula Ofek
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Lena Neufeld
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Adva Krivitsky
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Daniella Vaskovich-Koubi
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Ron Kleiner
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Pradip Dey
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Shani Koshrovski-Michael
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Noa Reisman
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel.
| |
Collapse
|
17
|
Yang J, Tang Z, Liu Y, Luo Z, Xiao Y, Zhang X. Comparison of chondro-inductivity between collagen and hyaluronic acid hydrogel based on chemical/physical microenvironment. Int J Biol Macromol 2021; 182:1941-1952. [PMID: 34062160 DOI: 10.1016/j.ijbiomac.2021.05.188] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 05/14/2021] [Accepted: 05/27/2021] [Indexed: 02/09/2023]
Abstract
Achieving chondrogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) successfully is crucial for cartilage regeneration. To date, various hydrogels with different chemical microenvironment have been used to modulate chondrogenic differentiation of BMSCs, especially collagen and hyaluronic acid hydrogel. However, the chondro-inductive ability of collagen and hyaluronic acid hydrogel has not been evaluated yet and the different chemical and physical microenvironment of these two hydrogels increase the difficulty of comparison. In this study, three different hydrogels based on collagen and hyaluronic acid (self-assembled collagen hydrogel (Col), self-assembled collagen hydrogel cross-linked with genipin (Cgp), and methacrylated hyaluronic acid hydrogel (HA)) were prepared and their chondro-inductive ability on the encapsulated BMSCs was evaluated. Col and Cgp have the same chemical composition and similar microstructure, but are different from HA, while Cgp and HA hydrogels have the same mechanical strength. It was found that chemical and physical microenvironments of the hydrogels combined to influence cell condensation. Thanks to cell condensation was more likely to occur in collagen hydrogels in the early stage, the cartilage-induced ability was in the order of Col > Cgp > HA. However, the severe shrinkage of Col and Cgp resulted in no enough space for cell proliferation within hydrogels in the later stage. In contrast, relatively stable physical microenvironment of HA helped to maintain continuous production of cartilage-related matrix in the later stage. Overall, these results revealed that the chondro-inductive ability of collagen and hyaluronic acid hydrogel with different chemical and physical microenvironment cannot be evaluated by a particular time period. However, it provided important information for optimization and design of the future hydrogels towards successful repair of articular cartilage.
Collapse
Affiliation(s)
- Jirong Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 61004, Sichuan, China; Research Center for Human Tissue and Organs Degeneration, Institute Biomedical and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangzhou, China
| | - Zizhao Tang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 61004, Sichuan, China
| | - Yifan Liu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 61004, Sichuan, China
| | - Zhaocong Luo
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 61004, Sichuan, China
| | - Yumei Xiao
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 61004, Sichuan, China.
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 61004, Sichuan, China
| |
Collapse
|
18
|
Emmert M, Somorowsky F, Ebert J, Görick D, Heyn A, Rosenberger E, Wahl M, Heinrich D. Modulation of Mammalian Cell Behavior by Nanoporous Glass. Adv Biol (Weinh) 2021; 5:e2000570. [PMID: 33960740 DOI: 10.1002/adbi.202000570] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 03/31/2021] [Indexed: 11/08/2022]
Abstract
The introduction of novel bioactive materials to manipulate living cell behavior is a crucial topic for biomedical research and tissue engineering. Biomaterials or surface patterns that boost specific cell functions can enable innovative new products in cell culture and diagnostics. This study investigates the influence of the intrinsically nano-patterned surface of nanoporous glass membranes on the behavior of mammalian cells. Three different cell lines and primary human mesenchymal stem cells (hMSCs) proliferate readily on nanoporous glass membranes with mean pore sizes between 10 and 124 nm. In both proliferation and mRNA expression experiments, L929 fibroblasts show a distinct trend toward mean pore sizes >80 nm. For primary hMSCs, excellent proliferation is observed on all nanoporous surfaces. hMSCs on samples with 17 nm pore size display increased expression of COL10, COL2A1, and SOX9, especially during the first two weeks of culture. In the upside down culture, SK-MEL-28 cells on nanoporous glass resist the gravitational force and proliferate well in contrast to cells on flat references. The effect of paclitaxel treatment of MDA-MB-321 breast cancer cells is already visible after 48 h on nanoporous membranes and strongly pronounced in comparison to reference samples, underlining the material's potential for functional drug screening.
Collapse
Affiliation(s)
- Martin Emmert
- Fraunhofer Institute for Silicate Research ISC, Neunerplatz 2, 97082, Würzburg, Germany.,Julius-Maximilians-Universität Würzburg, Chemical Technology of Material Synthesis, Röntgenring 11, 97070, Würzburg, Germany
| | - Ferdinand Somorowsky
- Fraunhofer Institute for Silicate Research ISC, Neunerplatz 2, 97082, Würzburg, Germany
| | - Jutta Ebert
- Fraunhofer Institute for Silicate Research ISC, Neunerplatz 2, 97082, Würzburg, Germany
| | - Dominik Görick
- Fraunhofer Institute for Silicate Research ISC, Neunerplatz 2, 97082, Würzburg, Germany
| | - Andreas Heyn
- Julius-Maximilians-Universität Würzburg, Chemical Technology of Material Synthesis, Röntgenring 11, 97070, Würzburg, Germany
| | - Eva Rosenberger
- Fraunhofer Institute for Silicate Research ISC, Neunerplatz 2, 97082, Würzburg, Germany
| | - Moritz Wahl
- Julius-Maximilians-Universität Würzburg, Chemical Technology of Material Synthesis, Röntgenring 11, 97070, Würzburg, Germany
| | - Doris Heinrich
- Fraunhofer Institute for Silicate Research ISC, Neunerplatz 2, 97082, Würzburg, Germany.,Leiden University, LION Leiden Institute of Physics, Niels Bohrweg 2, Leiden, 2333 CA, The Netherlands
| |
Collapse
|
19
|
Recent Developed Strategies for Enhancing Chondrogenic Differentiation of MSC: Impact on MSC-Based Therapy for Cartilage Regeneration. Stem Cells Int 2021; 2021:8830834. [PMID: 33824665 PMCID: PMC8007380 DOI: 10.1155/2021/8830834] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 02/20/2021] [Accepted: 03/04/2021] [Indexed: 12/19/2022] Open
Abstract
Articular cartilage is susceptible to damage, but its self-repair is hindered by its avascular nature. Traditional treatment methods are not able to achieve satisfactory repair effects, and the development of tissue engineering techniques has shed new light on cartilage regeneration. Mesenchymal stem cells (MSCs) are one of the most commonly used seed cells in cartilage tissue engineering. However, MSCs tend to lose their multipotency, and the composition and structure of cartilage-like tissues formed by MSCs are far from those of native cartilage. Thus, there is an urgent need to develop strategies that promote MSC chondrogenic differentiation to give rise to durable and phenotypically correct regenerated cartilage. This review provides an overview of recent advances in enhancement strategies for MSC chondrogenic differentiation, including optimization of bioactive factors, culture conditions, cell type selection, coculture, gene editing, scaffolds, and physical stimulation. This review will aid the further understanding of the MSC chondrogenic differentiation process and enable improvement of MSC-based cartilage tissue engineering.
Collapse
|
20
|
Thorp H, Kim K, Kondo M, Maak T, Grainger DW, Okano T. Trends in Articular Cartilage Tissue Engineering: 3D Mesenchymal Stem Cell Sheets as Candidates for Engineered Hyaline-Like Cartilage. Cells 2021; 10:cells10030643. [PMID: 33805764 PMCID: PMC7998529 DOI: 10.3390/cells10030643] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/05/2021] [Accepted: 03/10/2021] [Indexed: 02/07/2023] Open
Abstract
Articular cartilage defects represent an inciting factor for future osteoarthritis (OA) and degenerative joint disease progression. Despite multiple clinically available therapies that succeed in providing short term pain reduction and restoration of limited mobility, current treatments do not reliably regenerate native hyaline cartilage or halt cartilage degeneration at these defect sites. Novel therapeutics aimed at addressing limitations of current clinical cartilage regeneration therapies increasingly focus on allogeneic cells, specifically mesenchymal stem cells (MSCs), as potent, banked, and available cell sources that express chondrogenic lineage commitment capabilities. Innovative tissue engineering approaches employing allogeneic MSCs aim to develop three-dimensional (3D), chondrogenically differentiated constructs for direct and immediate replacement of hyaline cartilage, improve local site tissue integration, and optimize treatment outcomes. Among emerging tissue engineering technologies, advancements in cell sheet tissue engineering offer promising capabilities for achieving both in vitro hyaline-like differentiation and effective transplantation, based on controlled 3D cellular interactions and retained cellular adhesion molecules. This review focuses on 3D MSC-based tissue engineering approaches for fabricating “ready-to-use” hyaline-like cartilage constructs for future rapid in vivo regenerative cartilage therapies. We highlight current approaches and future directions regarding development of MSC-derived cartilage therapies, emphasizing cell sheet tissue engineering, with specific focus on regulating 3D cellular interactions for controlled chondrogenic differentiation and post-differentiation transplantation capabilities.
Collapse
Affiliation(s)
- Hallie Thorp
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112, USA; (H.T.); (M.K.); (D.W.G.)
- Department of Biomedical Engineering, University of Utah, 36 S Wasatch Dr, Salt Lake City, UT 84112, USA
| | - Kyungsook Kim
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112, USA; (H.T.); (M.K.); (D.W.G.)
- Correspondence: (K.K.); (T.O.); Tel.: +1-801-585-0070 (K.K. & T.O.); Fax: +1-801-581-3674 (K.K. & T.O.)
| | - Makoto Kondo
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112, USA; (H.T.); (M.K.); (D.W.G.)
| | - Travis Maak
- Department of Orthopaedic Surgery, University of Utah, 590 Wakara Way, Salt Lake City, UT 84108, USA;
| | - David W. Grainger
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112, USA; (H.T.); (M.K.); (D.W.G.)
- Department of Biomedical Engineering, University of Utah, 36 S Wasatch Dr, Salt Lake City, UT 84112, USA
| | - Teruo Okano
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112, USA; (H.T.); (M.K.); (D.W.G.)
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University, Wakamatsucho, 2−2, Shinjuku-ku, Tokyo 162-8480, Japan
- Correspondence: (K.K.); (T.O.); Tel.: +1-801-585-0070 (K.K. & T.O.); Fax: +1-801-581-3674 (K.K. & T.O.)
| |
Collapse
|
21
|
Helgeland E, Rashad A, Campodoni E, Goksøyr Ø, Pedersen TØ, Sandri M, Rosén A, Mustafa K. Dual-crosslinked 3D printed gelatin scaffolds with potential for temporomandibular joint cartilage regeneration. Biomed Mater 2021; 16. [DOI: 10.1088/1748-605x/abe6d9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 02/16/2021] [Indexed: 01/16/2023]
|
22
|
Celik C, Franco-Obregón A, Lee EH, Hui JH, Yang Z. Directionalities of magnetic fields and topographic scaffolds synergise to enhance MSC chondrogenesis. Acta Biomater 2021; 119:169-183. [PMID: 33130304 DOI: 10.1016/j.actbio.2020.10.039] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 10/15/2020] [Accepted: 10/26/2020] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cell (MSC) chondrogenesis is modulated by diverse biophysical cues. We have previously shown that brief, low-amplitude pulsed electromagnetic fields (PEMFs) differentially enhance MSC chondrogenesis in scaffold-free pellet cultures versus conventional tissue culture plastic (TCP), indicating an interplay between magnetism and micromechanical environment. Here, we examined the influence of PEMF directionality over the chondrogenic differentiation of MSCs laden on electrospun fibrous scaffolds of either random (RND) or aligned (ALN) orientations. Correlating MSCs' chondrogenic outcome to pFAK activation and YAP localisation, MSCs on the RND scaffolds experienced the least amount of resting mechanical stress and underwent greatest chondrogenic differentiation in response to brief PEMF exposure (10 min at 1 mT) perpendicular to the dominant plane of the scaffolds (Z-directed). By contrast, in MSC-impregnated RND scaffolds, greatest mitochondrial respiration resulted from X-directed PEMF exposure (parallel to the scaffold plane), and was associated with curtailed chondrogenesis. MSCs on TCP or the ALN scaffolds exhibited greater resting mechanical stress and accordingly, were unresponsive, or negatively responsive, to PEMF exposure from all directions. The efficacy of PEMF-induced MSC chondrogenesis is hence regulated in a multifaceted manner involving focal adhesion dynamics, as well as mitochondrial responses, culminating in a final cellular response. The combined contributions of micromechanical environment and magnetic field orientation hence will need to be considered when designing magnetic exposure paradigms.
Collapse
Affiliation(s)
- Cenk Celik
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228
| | - Alfredo Franco-Obregón
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228; BioIonic Currents Electromagnetic Pulsing Systems Laboratory, BICEPS, National University of Singapore, Singapore, 117599; Institute for Health Innovation & Technology, iHealthtech, National University of Singapore, Singapore, 117599; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, DSO (Kent Ridge) Building, #04-01, 27 Medical Drive, Singapore, 117593.
| | - Eng Hin Lee
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228; Tissue Engineering Program (NUSTEP), Life Sciences Institute, National University of Singapore, DSO (Kent Ridge) Building, #04-01, 27 Medical Drive, Singapore, 117510
| | - James Hp Hui
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228; Tissue Engineering Program (NUSTEP), Life Sciences Institute, National University of Singapore, DSO (Kent Ridge) Building, #04-01, 27 Medical Drive, Singapore, 117510
| | - Zheng Yang
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228; Tissue Engineering Program (NUSTEP), Life Sciences Institute, National University of Singapore, DSO (Kent Ridge) Building, #04-01, 27 Medical Drive, Singapore, 117510.
| |
Collapse
|
23
|
Effect of Nanostructured Scaffold on Human Adipose-Derived Stem Cells: Outcome of In Vitro Experiments. NANOMATERIALS 2020; 10:nano10091822. [PMID: 32932658 PMCID: PMC7558271 DOI: 10.3390/nano10091822] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/26/2020] [Accepted: 09/10/2020] [Indexed: 12/11/2022]
Abstract
This work is addressed to provide, by in vitro experiments, results on the repercussion that a nanostructured scaffold could have on viability, differentiation and secretion of bioactive factors of human adipose-derived stem cells (hASCs) when used in association to promote angiogenesis, a crucial condition to favour tissue regeneration. To achieve this aim, we evaluated cell viability and morphology by MTT (3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide) assay and microscopy analysis, respectively. We also investigated the expression of some of those genes involved in angiogenesis and differentiation processes utilizing quantitative polymerase chain reaction (qPCR), whereas the amounts of Vascular Endothelial Growth Factor A, Interleukin 6 and Fatty Acid-Binding Protein 4 secreted in the culture medium, were quantified by enzyme-linked immunosorbent assay (ELISA). Results suggested that, in the presence of the scaffold, cell proliferation and the exocytosis of factors involved in the angiogenesis process are reduced; by contrast, the expression of those genes involved in hASC differentiation appeared enhanced. To guarantee cell survival, the construct dimensions are, generally, smaller than clinically required. Furthermore, being the paracrine event the primary mechanism exerting the beneficial effects on injured tissues, the use of conditioned culture medium instead of cells may be convenient.
Collapse
|
24
|
Baracho Trindade Hill A, Speri Alves AA, da Silva Nunes Barreto R, Fernandes Bressan F, Miglino MA, Mansano Garcia J. Placental scaffolds have the ability to support adipose-derived cells differentiation into osteogenic and chondrogenic lineages. J Tissue Eng Regen Med 2020; 14:1661-1672. [PMID: 32893450 DOI: 10.1002/term.3124] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/29/2020] [Accepted: 08/25/2020] [Indexed: 12/23/2022]
Abstract
Prudent choices of cell sources and biomaterials, as well as meticulous cultivation of the tissue microenvironment, are essential to improving outcomes of tissue engineering treatments. With the goal of providing a high-quality alternative for bone and cartilage tissue engineering, we investigated the capability of bovine placental scaffolds to support adipose-derived cell differentiation into osteogenic and chondrogenic lineages. Decellularized bovine placenta, a high-quality scaffold with practical scalability, was chosen as the biomaterial due to its rich extracellular matrix, well-developed vasculature, high availability, low cost, and simplicity of collection. Adipose-derived cells were chosen as the cell source as they are easy to isolate, nontumorigenic, and flexibly differentiable. The bovine model was chosen for its advantages in translational medicine over the mouse model. When seeded onto the scaffolds, the isolated cells adhered to the scaffolds with cell projections, established cell-scaffold communication and proliferated while maintaining cell-cell communication. Throughout a 21-day culture period, osteogenically differentiated cells secreted mineralized matrix, and calcium deposits were observed throughout the scaffold. Under chondrogenic specific differentiation conditions, the cells modified their morphology from fibroblast-like to round cells and cartilage lacunas were observed as well as the deposit of cartilaginous matrix on the placental scaffolds. This experiment provides evidence, for the first time, that bovine placental scaffolds have the potential to support bovine mesenchymal stem cell adherence and differentiation into osteogenic and chondrogenic lineages. Therefore, the constructed material could be used for bone and cartilage tissue engineering.
Collapse
Affiliation(s)
- Amanda Baracho Trindade Hill
- Reproduction and Fertility Research Center, University of Montreal, Saint-Hyacinthe, QC, Canada.,Department of Preventive Veterinary Medicine and Animal Reproduction, São Paulo State University, Jaboticabal, Brazil
| | | | | | - Fabiana Fernandes Bressan
- Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, Brazil
| | - Maria Angelica Miglino
- School of Veterinary Medicine and Animal Sciences, University of São Paulo, São Paulo, Brazil
| | - Joaquim Mansano Garcia
- Department of Preventive Veterinary Medicine and Animal Reproduction, São Paulo State University, Jaboticabal, Brazil
| |
Collapse
|
25
|
Yang J, Xiao Y, Tang Z, Luo Z, Li D, Wang Q, Zhang X. The negatively charged microenvironment of collagen hydrogels regulates the chondrogenic differentiation of bone marrow mesenchymal stem cells in vitro and in vivo. J Mater Chem B 2020; 8:4680-4693. [PMID: 32391834 DOI: 10.1039/d0tb00172d] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The differentiation of bone marrow mesenchymal stem cells (BMSCs) into functional chondrocytes is crucial for successful cartilage tissue engineering. Since the extracellular matrix (ECM) microenvironment can regulate the behaviours of BMSCs and guide their differentiation, it is important to simulate the natural cartilage ECM to induce the chondrogenesis of BMSCs. As the most abundant protein in the ECM, collagen hydrogels were found to provide a structural and chemical microenvironment for natural cartilage, and regulate the chondrogenic differentiation of BMSCs. However, as the negatively charged ECM microenvironment is crucial for chondrogenesis and homeostasis within cells in cartilage tissue, the electrical properties of collagen hydrogels need to be further optimized. In this study, three collagen hydrogels with different electrical properties were fabricated using methacrylic anhydride (MA) and succinic anhydride (SA) modification. The collagen hydrogels had a similar composition, storage modulus and integral triple helix structure of collagen, but their different negatively charged microenvironments significantly impacted the hydrophilicity, protein diffusion and binding, and consequently influenced BMSC adhesion and spreading on the surface of the hydrogels. Moreover, the BMSCs encapsulated in the collagen hydrogels also demonstrated improved sGAG secretion and chondrogenic and integrin gene expression with the increased negative charge in vitro. Similar results were also observed in subcutaneous implantation in vivo, where higher secretions of sGAG, SOX9 and collagen type II proteins were found in the collagen hydrogels with higher negative charge. Together, our results demonstrated that more negative charges introduced into the collagen hydrogel microenvironment would enhance the chondrogenic differentiation of BMSCs in vitro and in vivo. This revealed that the electrical properties are an important consideration in designing future collagen hydrogels for cartilage regeneration.
Collapse
Affiliation(s)
- Jirong Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 61004, Sichuan, China.
| | | | | | | | | | | | | |
Collapse
|
26
|
Yang KC, Chen IH, Yang YT, Hsiao JK, Wang CC. Effects of scaffold geometry on chondrogenic differentiation of adipose-derived stem cells. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 110:110733. [DOI: 10.1016/j.msec.2020.110733] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 01/18/2020] [Accepted: 02/05/2020] [Indexed: 01/01/2023]
|
27
|
Piezoelectricity in the Intervertebral disc. J Biomech 2020; 102:109622. [DOI: 10.1016/j.jbiomech.2020.109622] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 12/18/2019] [Accepted: 01/06/2020] [Indexed: 12/17/2022]
|
28
|
Jeske SS, Theodoraki MN, Boelke E, Laban S, Brunner C, Rotter N, Jackson EK, Hoffmann TK, Schuler PJ. Adenosine production in mesenchymal stromal cells in relation to their developmental status. HNO 2020; 68:87-93. [PMID: 31915882 DOI: 10.1007/s00106-019-00805-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Mesenchymal stromal cells (MSC) are multipotent progenitor cells found in the tumor microenvironment. They have an innate and regulatory immune activity, and they are able to produce immunosuppressive adenosine (ADO) via their ectonucleotidases CD39 and CD73. The present study explores ADO metabolism of MSC in relation to their developmental status. METHODS We analyzed MSC (n = 6), chondrogenic progenitor cells (CPC, n = 8), and chondrocytes (n = 8) for surface markers by flow cytometry. The ability to hydrolyze ATP and to produce ADO was tested by luminescence assays and mass spectrometry. RESULTS Significant differences in the surface marker expression of MSC, CPC, and chondrocytes were seen. While the expression of CD73 was observed to be the same on all cell types, the expression of the ectonucleotidase CD39 was significantly increased on MSC. Consequently, production of ADO was most abundant in MSC as compared with chondrocytes and CPC. CONCLUSION Mesenchymal stromal cells are potent producers of ADO and are, therefore, able to increase immunosuppression. As MSC differentiate into chondrocytes, they lose this ability and may take on other functions.
Collapse
Affiliation(s)
- S S Jeske
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery, Ulm University Medical Center, Frauensteige 12, 89075, Ulm, Germany
| | - M N Theodoraki
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery, Ulm University Medical Center, Frauensteige 12, 89075, Ulm, Germany
| | - E Boelke
- Department of Radiotherapy and Radiooncology, Heinrich Heine University, Düsseldorf, Germany
| | - S Laban
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery, Ulm University Medical Center, Frauensteige 12, 89075, Ulm, Germany
| | - C Brunner
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery, Ulm University Medical Center, Frauensteige 12, 89075, Ulm, Germany
| | - N Rotter
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery, Mannheim University Medical Center, Mannheim, Germany
| | - E K Jackson
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - T K Hoffmann
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery, Ulm University Medical Center, Frauensteige 12, 89075, Ulm, Germany
| | - P J Schuler
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery, Ulm University Medical Center, Frauensteige 12, 89075, Ulm, Germany.
| |
Collapse
|
29
|
Drobnik J, Pietrucha K, Janczar K, Polis L, Polis B, Safandowska M, Szymański J. Intra-cerebral implantation of a variety of collagenous scaffolds with nervous embryonic cells. Exp Ther Med 2019; 18:4758-4764. [PMID: 31772645 PMCID: PMC6862020 DOI: 10.3892/etm.2019.8116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 08/27/2019] [Indexed: 12/28/2022] Open
Abstract
Collagenous scaffolds provide good conditions for embryonic nerve cell growth. The aim of the current study was to assess the brains reaction to the implantation of 3D sponge-shaped scaffolds. These scaffolds consisted of collagen (Col) and Col with chondroitin sulphate, which is modified by carbodiimide, or Col crosslinked with dialdehyde cellulose. The current study also evaluated the expression of integrins α2 and β1 in embryonic nerve cells. Embryonic nerve cells were isolated from the brains of rat embryos. Acellular scaffolds, or scaffolds populated with embryonic nerve cells, were implanted into the rats brain. The fibers of all the implanted scaffolds remained intact and served as a template for cell infiltration. The implants induced minimal to moderate inflammatory responses and minimal glial scar formations. Immunohistochemical studies did not indicate any microtubule-associated protein 2 or glial fibrillary acidic protein-positive cells inside the scaffolds. Acellular and cell-populated scaffolds yielded similar responses in the brain. The expression of integrin α2 and β1 was observed in embryonic nervous cells. TC-I15, the integrin α2β1 inhibitor, was not demonstrated to modify cell entrapment within the collagenous scaffolds. All applied scaffolds were well tolerated by the tissue and were indicated to support blood vessel formation. Therefore, all tested biomaterials are recommended for further studies. Additional chemical modifications of the material are suggested to protect the seeded cells from apoptosis after implantation into the brain.
Collapse
Affiliation(s)
- Jacek Drobnik
- Laboratory of Connective Tissue Metabolism, Department of Pathophysiology, Medical University of Lodz, 90-136 Lodz, Poland
| | - Krystyna Pietrucha
- Department of Material and Commodity Sciences and Textile Metrology, Lodz University of Technology, 90-924 Lodz, Poland
| | - Karolina Janczar
- Department of Pathomorphology, Medical University of Lodz, 92-213 Lodz, Poland
| | - Lech Polis
- Department of Neurosurgery, Polish Mothers' Memorial Hospital-Research Institute, 93-338 Lodz, Poland
| | - Bartosz Polis
- Department of Neurosurgery, Polish Mothers' Memorial Hospital-Research Institute, 93-338 Lodz, Poland
| | - Marta Safandowska
- Department of Material and Commodity Sciences and Textile Metrology, Lodz University of Technology, 90-924 Lodz, Poland
| | - Jacek Szymański
- Central Scientific Laboratory, Medical University of Lodz, 92-215 Lodz, Poland
| |
Collapse
|
30
|
Li YY, Lam KL, Chen AD, Zhang W, Chan BP. Collagen microencapsulation recapitulates mesenchymal condensation and potentiates chondrogenesis of human mesenchymal stem cells – A matrix-driven in vitro model of early skeletogenesis. Biomaterials 2019; 213:119210. [DOI: 10.1016/j.biomaterials.2019.05.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 04/28/2019] [Accepted: 05/10/2019] [Indexed: 01/01/2023]
|
31
|
Kavand H, van Lintel H, Renaud P. Efficacy of pulsed electromagnetic fields and electromagnetic fields tuned to the ion cyclotron resonance frequency of Ca 2+ on chondrogenic differentiation. J Tissue Eng Regen Med 2019; 13:799-811. [PMID: 30793837 DOI: 10.1002/term.2829] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 02/05/2019] [Accepted: 02/21/2019] [Indexed: 12/17/2022]
Abstract
Previous studies provide strong evidence for the therapeutic effect of electromagnetic fields (EMFs) on different tissues including cartilage. Diverse exposure parameters applied in scientific reports and the unknown interacting mechanism of EMF with biological systems make EMF studies challenging. In 1985, Liboff proposed that when magnetic fields are tuned to the cyclotron resonance frequencies of critical ions, the motion of ions through cell membranes is enhanced, and thus biological effects appear. Such exposure system consists of a weak alternating magnetic field (B1 ) in the presence of a static magnetic field (B0 ) and depends on the relationship between the magnitudes of B0 and B1 and the angular frequency Ω. The purpose of the present study is to determine the chondrogenic potential of EMF with regards to pulsed EMF (PEMF) and the ion cyclotron resonance (ICR) theory. We used different stimulating systems to generate EMFs in which cells are either stimulated with ubiquitous PEMF parameters, frequently reported, or parameters tuned to satisfy the ICR for Ca2+ (including negative and positive control groups). Chondrogenesis was analysed after 3 weeks of treatment. Cell stimulation under the ICR condition showed positive results in the context of glycosaminoglycans and type II collagen synthesis. In contrast, the other electromagnetically stimulated groups showed no changes compared with the control groups. Furthermore, gene expression assays revealed an increase in the expression of chondrogenic markers (COL2A1, SOX9, and ACAN) in the ICR group. These results suggest that the Ca2+ ICR condition can be an effective factor in inducing chondrogenesis.
Collapse
Affiliation(s)
- Hanie Kavand
- Microsystems Laboratory, Institute of Microengineering, School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Harald van Lintel
- Microsystems Laboratory, Institute of Microengineering, School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Philippe Renaud
- Microsystems Laboratory, Institute of Microengineering, School of Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
32
|
Liu J, Wang X, Lu G, Tang JZ, Wang Y, Zhang B, Sun Y, Lin H, Wang Q, Liang J, Fan Y, Zhang X. Bionic cartilage acellular matrix microspheres as a scaffold for engineering cartilage. J Mater Chem B 2019; 7:640-650. [DOI: 10.1039/c8tb02999g] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Bionic cartilage acellular matrix microspheres (BCAMMs) made from decelluarized bionic cartilage microspheres (BCMs).
Collapse
|
33
|
Irawan V, Sung TC, Higuchi A, Ikoma T. Collagen Scaffolds in Cartilage Tissue Engineering and Relevant Approaches for Future Development. Tissue Eng Regen Med 2018; 15:673-697. [PMID: 30603588 PMCID: PMC6250655 DOI: 10.1007/s13770-018-0135-9] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 05/30/2018] [Accepted: 06/15/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Cartilage tissue engineering (CTE) aims to obtain a structure mimicking native cartilage tissue through the combination of relevant cells, three-dimensional scaffolds, and extraneous signals. Implantation of 'matured' constructs is thus expected to provide solution for treating large injury of articular cartilage. Type I collagen is widely used as scaffolds for CTE products undergoing clinical trial, owing to its ubiquitous biocompatibility and vast clinical approval. However, the long-term performance of pure type I collagen scaffolds would suffer from its limited chondrogenic capacity and inferior mechanical properties. This paper aims to provide insights necessary for advancing type I collagen scaffolds in the CTE applications. METHODS Initially, the interactions of type I/II collagen with CTE-relevant cells [i.e., articular chondrocytes (ACs) and mesenchymal stem cells (MSCs)] are discussed. Next, the physical features and chemical composition of the scaffolds crucial to support chondrogenic activities of AC and MSC are highlighted. Attempts to optimize the collagen scaffolds by blending with natural/synthetic polymers are described. Hybrid strategy in which collagen and structural polymers are combined in non-blending manner is detailed. RESULTS Type I collagen is sufficient to support cellular activities of ACs and MSCs; however it shows limited chondrogenic performance than type II collagen. Nonetheless, type I collagen is the clinically feasible option since type II collagen shows arthritogenic potency. Physical features of scaffolds such as internal structure, pore size, stiffness, etc. are shown to be crucial in influencing the differentiation fate and secreting extracellular matrixes from ACs and MSCs. Collagen can be blended with native or synthetic polymer to improve the mechanical and bioactivities of final composites. However, the versatility of blending strategy is limited due to denaturation of type I collagen at harsh processing condition. Hybrid strategy is successful in maximizing bioactivity of collagen scaffolds and mechanical robustness of structural polymer. CONCLUSION Considering the previous improvements of physical and compositional properties of collagen scaffolds and recent manufacturing developments of structural polymer, it is concluded that hybrid strategy is a promising approach to advance further collagen-based scaffolds in CTE.
Collapse
Affiliation(s)
- Vincent Irawan
- Department of Materials Science and Engineering, Tokyo Institute of Technology, 2 Chome-12-1, Meguro-ku, Tokyo, 152-8550 Japan
| | - Tzu-Cheng Sung
- Department of Chemical and Materials Engineering, National Central University, No. 300 Jung Da Rd., Chung-Li, Taoyuan, 320 Taiwan
| | - Akon Higuchi
- Department of Chemical and Materials Engineering, National Central University, No. 300 Jung Da Rd., Chung-Li, Taoyuan, 320 Taiwan
| | - Toshiyuki Ikoma
- Department of Materials Science and Engineering, Tokyo Institute of Technology, 2 Chome-12-1, Meguro-ku, Tokyo, 152-8550 Japan
| |
Collapse
|
34
|
Wen J, Zhao Z, Tong R, Huang L, Miao Y, Wu J. Prussian Blue Nanoparticle-Labeled Mesenchymal Stem Cells: Evaluation of Cell Viability, Proliferation, Migration, Differentiation, Cytoskeleton, and Protein Expression In Vitro. NANOSCALE RESEARCH LETTERS 2018; 13:329. [PMID: 30350300 PMCID: PMC6197343 DOI: 10.1186/s11671-018-2730-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 09/24/2018] [Indexed: 02/05/2023]
Abstract
Mesenchymal stem cells (MSCs) have been used for the treatment of various human diseases. To better understand the mechanism of this action and the fate of these cells, magnetic resonance imaging (MRI) has been used for the tracking of transplanted stem cells. Prussian blue nanoparticles (PBNPs) have been demonstrated to have the ability of labeling cells to visualize them as an effective MRI contrast agent. In this study, we aimed to investigate the efficiency and biological effects of labeled MSCs using PBNPs. We first synthesized and characterized the PBNPs. Then, iCELLigence real-time cell analysis system revealed that PBNPs did not significantly alter cell viability, proliferation, and migration activity in PBNP-labeled MSCs. Oil Red O staining and Alizarin Red staining revealed that labeled MSCs also have a normal differentiation capacity. Phalloidin staining showed no negative effect of PBNPs on the cytoskeleton. Western blot analysis indicated that PBNPs also did not change the expression of β-catenin and vimentin of MSCs. In vitro MRI, the pellets of the MSCs incubated with PBNPs showed a clear MRI signal darkening effect. In conclusion, PBNPs can be effectively used for the labeling of MSCs and will not influence the biological characteristics of MSCs.
Collapse
Affiliation(s)
- Jirui Wen
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, No17, 3rd section, Renmin Nanlu Road, Chengdu, 610041, Sichuan, China.,Department of Gynecology, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Chengdu, China
| | - Zhiwei Zhao
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, No17, 3rd section, Renmin Nanlu Road, Chengdu, 610041, Sichuan, China
| | - Ruijie Tong
- College of Pharmaceutical and Biological Engineering, Shenyang University of Chemical Technology, Shenyang, China
| | - Liwei Huang
- West China School of Stomatology Medicine, Sichuan University, Chengdu, China
| | - Yali Miao
- Department of Gynecology, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Chengdu, China. .,Deep Undergroud Space Medical Center, West China Hospital, Sichuan University, No.17, 3rd Section, Renmin Nanlu Road, Chengdu, 610041, Sichuan, China.
| | - Jiang Wu
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, No17, 3rd section, Renmin Nanlu Road, Chengdu, 610041, Sichuan, China. .,Deep Undergroud Space Medical Center, West China Hospital, Sichuan University, No.17, 3rd Section, Renmin Nanlu Road, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
35
|
3D Bone Biomimetic Scaffolds for Basic and Translational Studies with Mesenchymal Stem Cells. Int J Mol Sci 2018; 19:ijms19103150. [PMID: 30322134 PMCID: PMC6213614 DOI: 10.3390/ijms19103150] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 10/04/2018] [Accepted: 10/10/2018] [Indexed: 12/22/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are recognized as an attractive tool owing to their self-renewal and differentiation capacity, and their ability to secrete bioactive molecules and to regulate the behavior of neighboring cells within different tissues. Accumulating evidence demonstrates that cells prefer three-dimensional (3D) to 2D culture conditions, at least because the former are closer to their natural environment. Thus, for in vitro studies and in vivo utilization, great effort is being dedicated to the optimization of MSC 3D culture systems in view of achieving the intended performance. This implies understanding cell–biomaterial interactions and manipulating the physicochemical characteristics of biomimetic scaffolds to elicit a specific cell behavior. In the bone field, biomimetic scaffolds can be used as 3D structures, where MSCs can be seeded, expanded, and then implanted in vivo for bone repair or bioactive molecules release. Actually, the union of MSCs and biomaterial has been greatly improving the field of tissue regeneration. Here, we will provide some examples of recent advances in basic as well as translational research about MSC-seeded scaffold systems. Overall, the proliferation of tools for a range of applications witnesses a fruitful collaboration among different branches of the scientific community.
Collapse
|
36
|
Gullotta F, Izzo D, Scalera F, Palazzo B, Martin I, Sannino A, Gervaso F. Biomechanical evaluation of hMSCs-based engineered cartilage for chondral tissue regeneration. J Mech Behav Biomed Mater 2018; 86:294-304. [DOI: 10.1016/j.jmbbm.2018.06.040] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 06/18/2018] [Accepted: 06/25/2018] [Indexed: 01/22/2023]
|
37
|
Huang N, Li CW, Chan BP. Multiphoton 3D Microprinting of Protein Micropatterns with Spatially Controlled Heterogeneity - A Platform for Single Cell Matrix Niche Studies. ACTA ACUST UNITED AC 2018. [DOI: 10.1002/adbi.201800053] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Nan Huang
- Tissue Engineering Laboratory; Department of Mechanical Engineering; The University of Hong Kong; Pokfulam Road Hong Kong Special Administrative Region China
| | - Chuen Wai Li
- Tissue Engineering Laboratory; Department of Mechanical Engineering; The University of Hong Kong; Pokfulam Road Hong Kong Special Administrative Region China
| | - Barbara Pui Chan
- Tissue Engineering Laboratory; Department of Mechanical Engineering; The University of Hong Kong; Pokfulam Road Hong Kong Special Administrative Region China
| |
Collapse
|
38
|
Thomas D, O'Brien T, Pandit A. Toward Customized Extracellular Niche Engineering: Progress in Cell-Entrapment Technologies. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:1703948. [PMID: 29194781 DOI: 10.1002/adma.201703948] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Revised: 09/12/2017] [Indexed: 06/07/2023]
Abstract
The primary aim in tissue engineering is to repair, replace, and regenerate dysfunctional tissues to restore homeostasis. Cell delivery for repair and regeneration is gaining impetus with our understanding of constructing tissue-like environments. However, the perpetual challenge is to identify innovative materials or re-engineer natural materials to model cell-specific tissue-like 3D modules, which can seamlessly integrate and restore functions of the target organ. To devise an optimal functional microenvironment, it is essential to define how simple is complex enough to trigger tissue regeneration or restore cellular function. Here, the purposeful transition of cell immobilization from a cytoprotection point of view to that of a cell-instructive approach is examined, with advances in the understanding of cell-material interactions in a 3D context, and with a view to further application of the knowledge for the development of newer and complex hierarchical tissue assemblies for better examination of cell behavior and offering customized cell-based therapies for tissue engineering.
Collapse
Affiliation(s)
- Dilip Thomas
- Centre for Research in Medical Devices (CÚRAM), National University of Ireland Galway, Galway, Ireland
- Regenerative Medicine Institute (REMEDI), National University of Ireland Galway, Galway, Ireland
- Cardiovascular Institute, Stanford University, Palo Alto, CA, 94305, USA
| | - Timothy O'Brien
- Regenerative Medicine Institute (REMEDI), National University of Ireland Galway, Galway, Ireland
| | - Abhay Pandit
- Centre for Research in Medical Devices (CÚRAM), National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
39
|
Ni Y, Tang Z, Yang J, Gao Y, Lin H, Guo L, Zhang K, Zhang X. Collagen structure regulates MSCs behavior by MMPs involved cell–matrix interactions. J Mater Chem B 2018; 6:312-326. [DOI: 10.1039/c7tb02377d] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Various scaffolds have been studied in the formation of cell niches and regulation of mesenchymal stem cells (MSCs) behaviors.
Collapse
Affiliation(s)
- Yilu Ni
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| | - Zhurong Tang
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| | - Jirong Yang
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| | - Yongli Gao
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| | - Hai Lin
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| | - Likun Guo
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| | - Kai Zhang
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials
- Sichuan University
- Chengdu 610064
- China
| |
Collapse
|
40
|
Zhang Q, Lin S, Zhang T, Tian T, Ma Q, Xie X, Xue C, Lin Y, Zhu B, Cai X. Curved microstructures promote osteogenesis of mesenchymal stem cells via the RhoA/ROCK pathway. Cell Prolif 2017; 50:e12356. [PMID: 28714177 PMCID: PMC6529063 DOI: 10.1111/cpr.12356] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVES Cells in the osteon reside in a curved space, accordingly, the curvature of the microenvironment is an important geometric feature in bone formation. However, it is not clear how curved microstructures affect cellular behaviour in bone tissue. MATERIALS AND METHODS Rat primary bone marrow mesenchymal stem cells (BMSCs) on wavy microgrooves were exposed to PDMS substrates with various curvatures to investigate alterations in cellular morphology and osteogenic differentiation. Additionally, the expression levels of RhoA and its effectors were examined by immunofluorescence and quantitative PCR to determine the mechanisms of curvature-dependent osteogenic differentiation. RESULTS Wavy microgrooves caused dramatic nuclear distortion and cytoskeletal remodelling. We detected a noticeable increase in the expression of osteogenic-related genes in BMSCs in wavy microgroove groups, and the maximum expression was observed in the high curvature group. Moreover, immunofluorescent staining and quantitative RT-PCR results for RhoA and its effectors showed that the RhoA/ROCK signalling pathway is associated with curvature-dependent osteogenic differentiation. CONCLUSIONS Our results illustrated that curved microstructures could promote BMSC differentiation to the osteogenic lineage, and the osteogenic effects of higher curvature are more obvious. Wavy microstructures could also influence the RhoA/ROCK pathway. Accordingly, curved microstructures may be useful in bone tissue engineering.
Collapse
Affiliation(s)
- Qi Zhang
- State Key Laboratory of Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengdu610041China
| | - Shiyu Lin
- State Key Laboratory of Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengdu610041China
| | - Tao Zhang
- State Key Laboratory of Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengdu610041China
| | - Taoran Tian
- State Key Laboratory of Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengdu610041China
| | - Quanquan Ma
- State Key Laboratory of Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengdu610041China
| | - Xueping Xie
- State Key Laboratory of Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengdu610041China
| | - Changyue Xue
- State Key Laboratory of Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengdu610041China
| | - Yunfeng Lin
- State Key Laboratory of Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengdu610041China
| | - Bofeng Zhu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine ResearchCollege of StomatologyXi'an Jiaotong UniversityXianChina
- Clinical Research Center of Shaanxi Province for Dental and Maxillofacial DiseasesCollege of StomatologyXi'an Jiaotong UniversityXianChina
| | - Xiaoxiao Cai
- State Key Laboratory of Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengdu610041China
| |
Collapse
|
41
|
Li X, Teng Y, Liu J, Lin H, Fan Y, Zhang X. Chondrogenic differentiation of BMSCs encapsulated in chondroinductive polysaccharide/collagen hybrid hydrogels. J Mater Chem B 2017; 5:5109-5119. [PMID: 32264096 DOI: 10.1039/c7tb01020f] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Although BMSC-based therapy is one of the most front-line technologies for cartilage repair, it is still a big challenge to attain ideal niches for BMSC chondrogenic differentiation. In this study, we developed hyaluronate and chondroitin sulfate derivatives to prepare covalently crosslinked polysaccharide hydrogels. Based on these binary hydrogels, collagen was added to prepare ternary hybrid hydrogels and its effect on encapsulated BMSCs was studied. After culturing with different cell densities in vitro without the addition of growth factors for 3 weeks, the chondrogenesis of BMSCs was evaluated by CLSM, mechanical testing, histological staining, immunohistochemical staining and gene expression. The results indicated that BMSCs in high cell density (50 million per mL) cell-laden constructs had a more obvious chondrogenic phenotype than those in low cell density ones (5 million per mL). However, the components of hydrogels had a significant influence on chondrogenic differentiation. With the addition of collagen, the BMSCs in ternary hybrid hydrogels showed more significant chondrogenesis, possessing with more amounts of secreted glycosaminoglycans (GAGs) and type II collagen deposition, higher mechanical properties and chondrogenic gene expression over 3 weeks of culture in vitro. It can be concluded that the bioactive collagen is beneficial to the chondrogenesis of BMSCs. This hybrid hydrogels deserve further studies to have a prospective application in tissue engineering for cartilage defect repair.
Collapse
Affiliation(s)
- Xiupeng Li
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu 610064, China.
| | | | | | | | | | | |
Collapse
|
42
|
Bierbaum S, Hintze V, Scharnweber D. 2.8 Artificial Extracellular Matrices to Functionalize Biomaterial Surfaces ☆. COMPREHENSIVE BIOMATERIALS II 2017:147-178. [DOI: 10.1016/b978-0-12-803581-8.10206-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
43
|
Hasturk O, Sivas A, Karasozen B, Demirci U, Hasirci N, Hasirci V. Quantification of Type, Timing, and Extent of Cell Body and Nucleus Deformations Caused by the Dimensions and Hydrophilicity of Square Prism Micropillars. Adv Healthc Mater 2016; 5:2972-2982. [PMID: 27925459 DOI: 10.1002/adhm.201600857] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 09/21/2016] [Indexed: 01/30/2023]
Abstract
Novel digital analysis strategies are developed for the quantification of changes in the cytoskeletal and nuclear morphologies of mesenchymal stem cells cultured on micropillars. Severe deformations of nucleus and distinct conformational changes of cell body ranging from extensive elongation to branching are visualized and quantified. These deformations are caused mainly by the dimensions and hydrophilicity of the micropillars.
Collapse
Affiliation(s)
- Onur Hasturk
- Graduate Department of Biotechnology; Middle East Technical University (METU); Ankara 06800 Turkey
- BIOMATEN; Center of Excellence in Biomaterials and Tissue Engineering; Middle East Technical University (METU); Ankara 06800 Turkey
| | - Abdullah Sivas
- Institute of Applied Mathematics; Middle East Technical University (METU); Ankara 06800 Turkey
| | - Bulent Karasozen
- Institute of Applied Mathematics; Middle East Technical University (METU); Ankara 06800 Turkey
| | - Utkan Demirci
- Bio-Acoustic-MEMs in Medicine (BAMM) Laboratory; Stanford School of Medicine; Palo Alto CA 94394 USA
| | - Nesrin Hasirci
- Graduate Department of Biotechnology; Middle East Technical University (METU); Ankara 06800 Turkey
- BIOMATEN; Center of Excellence in Biomaterials and Tissue Engineering; Middle East Technical University (METU); Ankara 06800 Turkey
- Department of Chemistry; Middle East Technical University (METU); Ankara 06800 Turkey
| | - Vasif Hasirci
- Graduate Department of Biotechnology; Middle East Technical University (METU); Ankara 06800 Turkey
- BIOMATEN; Center of Excellence in Biomaterials and Tissue Engineering; Middle East Technical University (METU); Ankara 06800 Turkey
- Department of Biological Sciences; Middle East Technical University (METU); Ankara 06800 Turkey
| |
Collapse
|
44
|
Wang J, Tao Y, Zhou X, Li H, Liang C, Li F, Chen QX. The potential of chondrogenic pre-differentiation of adipose-derived mesenchymal stem cells for regeneration in harsh nucleus pulposus microenvironment. Exp Biol Med (Maywood) 2016; 241:2104-2111. [PMID: 27488396 DOI: 10.1177/1535370216662362] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Recent studies indicated that cell-based therapy could be a promising approach to treat intervertebral disc degeneration. Though the harsh microenvironment in disc is still challenging to implanted cells, it could be overcome by pre-conditioning graft cells before transplantation, suggested by previous literatures. Therefore, we designed this study to identify the potential effect of chondrogenic pre-differentiation on adipose-derived mesenchymal stem cells in intervertebral disc-like microenvironment, characterized by limited nutrition, acidic, and high osmosis in vitro. Adipose-derived mesenchymal stem cells of rat were divided into five groups, embedded in type II collagen scaffold, and cultured in chondrogenic differentiation medium for 0, 3, 7, 10, and 14 days. Then, the adipose-derived mesenchymal stem cells were implanted and cultured in intervertebral disc-like condition. The proliferation and differentiation of adipose-derived mesenchymal stem cells were evaluated by cell counting kit-8 test, real-time quantitative polymerase chain reaction, and Western blotting and immunofluorescence analysis. Analyzed by the first week in intervertebral disc-like condition, the results showed relatively greater proliferative capability and extracellular matrix synthesis ability of the adipose-derived mesenchymal stem cells pre-differentiated for 7 and 10 days than the control. We concluded that pre-differentiation of rat adipose-derived mesenchymal stem cells in chondrogenic culture medium for 7 to 10 days could promote the regeneration effect of adipose-derived mesenchymal stem cells in intervertebral disc-like condition, and the pre-differentiated cells could be a promising cell source for disc regeneration medicine.
Collapse
Affiliation(s)
- Jingkai Wang
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Yiqing Tao
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Xiaopeng Zhou
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Hao Li
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Chengzhen Liang
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Fangcai Li
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Qi-Xin Chen
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| |
Collapse
|
45
|
Mamidi MK, Das AK, Zakaria Z, Bhonde R. Mesenchymal stromal cells for cartilage repair in osteoarthritis. Osteoarthritis Cartilage 2016; 24:1307-16. [PMID: 26973328 DOI: 10.1016/j.joca.2016.03.003] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 02/09/2016] [Accepted: 03/03/2016] [Indexed: 02/08/2023]
Abstract
Treatment for articular cartilage damage is quite challenging as it shows limited repair and regeneration following injury. Non-operative and classical surgical techniques are inefficient in restoring normal anatomy and function of cartilage in osteoarthritis (OA). Thus, investigating new and effective strategies for OA are necessary to establish feasible therapeutic solutions. The emergence of the new discipline of regenerative medicine, having cell-based therapy as its primary focus, may enable us to achieve repair and restore the damaged articular cartilage. This review describes progress and development of employing mesenchymal stromal cell (MSC)-based therapy as a promising alternative for OA treatment. The objective of this review is to first, discuss how in vitro MSC chondrogenic differentiation mimics in vivo embryonic cartilage development, secondly, to describe various chondrogenic differentiation strategies followed by pre-clinical and clinical studies demonstrating their feasibility and efficacy. However, several challenges need to be tackled before this research can be translated to the clinics. In particular, better understanding of the post-transplanted cell behaviour and learning to enhance their potency in the disease microenvironment is essential. Final objective is to underscore the importance of isolation, storage, cell shipment, route of administration, optimum dosage and control batch to batch variations to realise the full potential of MSCs in OA clinical trials.
Collapse
Affiliation(s)
- M K Mamidi
- School of Regenerative Medicine, Manipal University, Bangalore 560065, India
| | - A K Das
- Department of Surgery, Taylor's University School of Medicine, Sungai Buloh Hospital, Selangor, Malaysia
| | - Z Zakaria
- Hematology Unit, Cancer Research Centre, Institute for Medical Research, Jalan Pahang, 50588 Kuala Lumpur, Malaysia
| | - R Bhonde
- School of Regenerative Medicine, Manipal University, Bangalore 560065, India.
| |
Collapse
|
46
|
Mesenchymal Stem Cells Increase Neo-Angiogenesis and Albumin Production in a Liver Tissue-Engineered Engraftment. Int J Mol Sci 2016; 17:374. [PMID: 26985891 PMCID: PMC4813233 DOI: 10.3390/ijms17030374] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 02/12/2016] [Accepted: 03/01/2016] [Indexed: 12/16/2022] Open
Abstract
The construction of a three-dimensional (3D) liver tissue is limited by many factors; one of them is the lack of vascularization inside the tissue-engineered construct. An engineered liver pocket-scaffold able to increase neo-angiogenesis in vivo could be a solution to overcome these limitations. In this work, a hyaluronan (HA)-based scaffold enriched with human mesenchymal stem cells (hMSCs) and rat hepatocytes was pre-conditioned in a bioreactor system, then implanted into the liver of rats. Angiogenesis and hepatocyte metabolic functions were monitored. The formation of a de novo vascular network within the HA-based scaffold, as well as an improvement in albumin production by the implanted hepatocytes, were detected. The presence of hMSCs in the HA-scaffold increased the concentration of growth factors promoting angiogenesis inside the graft. This event ensured a high blood vessel density, coupled with a support to metabolic functions of hepatocytes. All together, these results highlight the important role played by stem cells in liver tissue-engineered engraftment.
Collapse
|
47
|
Protease inhibitors enhance extracellular collagen fibril deposition in human mesenchymal stem cells. Stem Cell Res Ther 2015; 6:197. [PMID: 26466582 PMCID: PMC4606504 DOI: 10.1186/s13287-015-0191-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 08/20/2015] [Accepted: 09/22/2015] [Indexed: 11/10/2022] Open
Abstract
INTRODUCTION Collagen is a widely used naturally occurring biomaterial for scaffolding, whereas mesenchymal stem cells (MSCs) represent a promising cell source in tissue engineering and regenerative medicine. It is generally known that cells are able to remodel their environment by simultaneous degradation of the scaffolds and deposition of newly synthesized extracellular matrix. Nevertheless, the interactions between MSCs and collagen biomaterials are poorly known, and the strategies enhancing the extracellular matrix deposition are yet to be defined. In this study, we aim to investigate the fate of collagen when it is in contact with MSCs and hypothesize that protease inhibition will enhance their extracellular deposition of collagen fibrils. METHODS Specifically, human MSCs (hMSCs) were exposed to fluorescence-labeled collagen with and without intracellular or extracellular protease inhibitors (or both) before tracing the collagen at both intracellular and extracellular spaces. RESULTS Collagen were internalized by hMSCs and degraded intracellularly in lysosomes. In the presence of protease inhibitors, both intracellular collagen fibril growth and extracellular deposition of collagen fibrils were enhanced. Moreover, protease inhibitors work synergistically with ascorbic acid, a well-known matrix deposition-enhancing reagent, in further enhancing collagen fibril deposition at the extracellular space. CONCLUSION These findings provide a better understanding of the interactions between hMSCs and collagen biomaterials and suggest a method to manipulate matrix remodeling and deposition of hMSCs, contributing to better scaffolding for tissue engineering and regenerative medicine.
Collapse
|
48
|
LeBlon CE, Casey ME, Fodor CR, Zhang T, Zhang X, Jedlicka SS. Correlation between in vitro expansion-related cell stiffening and differentiation potential of human mesenchymal stem cells. Differentiation 2015; 90:1-15. [PMID: 26381795 DOI: 10.1016/j.diff.2015.08.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 08/10/2015] [Accepted: 08/20/2015] [Indexed: 12/28/2022]
Abstract
Human mesenchymal stem cells (hMSCs) are an attractive cell source for tissue regeneration, given their self-renewal and multilineage potential. However, they are present in only small percentages in human bone marrow, and are generally propagated in vitro prior to downstream use. Previous work has shown that hMSC propagation can lead to alterations in cell behavior and differentiation potency, yet optimization of differentiation based on starting cell elastic modulus is an area still under investigation. To further advance the knowledge in this field, hMSCs were cultured and routinely passaged on tissue-culture polystyrene to investigate the correlation between cell stiffening and differentiation potency during in vitro aging. Local cell elastic modulus was measured at every passage using atomic force microscopy indentation. At each passage, cells were induced to differentiate down myogenic and osteogenic paths. Cells induced to differentiate, as well as undifferentiated cells were assessed for gene and protein expression using quantitative polymerase chain reaction and immunofluorescent staining, respectively, for osteogenic and myogenic markers. Myogenic and osteogenic cell potential are highly reliant on the elastic modulus of the starting cell population (of undifferentiated cells), and this potential appears to peak when the innate cell elastic modulus is close to that of differentiated tissue. However, the latent expression of the same markers in undifferentiated cells also appears to undergo a correlative relationship with cell elastic modulus, indicating some endogenous effects of cell elastic modulus and gene/protein expression. Overall, this study correlates age-related changes with regards to innate cell stiffening and gene/protein expression in commercial hMSCs, providing some guidance as to maintenance and future use of hMSCs in future tissue engineering applications.
Collapse
Affiliation(s)
- Courtney E LeBlon
- Mechanical Engineering & Mechanics, Packard Laboratory, Lehigh University, 19 Memorial Drive, Bethlehem, PA 18015, United States
| | - Meghan E Casey
- Bioengineering Program, Lehigh University, 111 Research Drive, Iacocca Hall, Bethlehem, PA 18015, United States
| | - Caitlin R Fodor
- Bioengineering Program, Lehigh University, 111 Research Drive, Iacocca Hall, Bethlehem, PA 18015, United States
| | - Tony Zhang
- Bioengineering Program, Lehigh University, 111 Research Drive, Iacocca Hall, Bethlehem, PA 18015, United States
| | - Xiaohui Zhang
- Mechanical Engineering & Mechanics, Packard Laboratory, Lehigh University, 19 Memorial Drive, Bethlehem, PA 18015, United States; Bioengineering Program, Lehigh University, 111 Research Drive, Iacocca Hall, Bethlehem, PA 18015, United States
| | - Sabrina S Jedlicka
- Bioengineering Program, Lehigh University, 111 Research Drive, Iacocca Hall, Bethlehem, PA 18015, United States; Materials Science and Engineering, Whitaker Laboratory, Lehigh University, 5 East Packer Ave., Bethlehem, PA 18015, United States; Center for Advanced Materials & Nanotechnology, Whitaker Laboratory, Lehigh University, 5 East Packer Ave., Bethlehem, PA 18015, United States.
| |
Collapse
|