1
|
Kao YC, Yang PC, Lin YP, Chen GH, Liu SW, Ho CH, Huang SC, Lee PY, Chen L, Huang CC. Tailoring the therapeutic potential of stem cell spheroid-derived decellularized ECM through post-decellularization BDNF incorporation to enhance brain repair. Biomaterials 2025; 321:123332. [PMID: 40220567 DOI: 10.1016/j.biomaterials.2025.123332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 04/06/2025] [Accepted: 04/07/2025] [Indexed: 04/14/2025]
Abstract
Decellularized extracellular matrix (dECM) from tissues has significant therapeutic potential but is limited by its rigid molecular composition and reliance on post-decellularization modifications to tailor its functionality. Harsh decellularization processes often result in substantial glycosaminoglycan (GAG) loss, impairing natural growth factor incorporation and necessitating chemical modifications that complicate processing and limit clinical translation. To address these challenges, we developed mesenchymal stem cell (MSC) spheroid-derived three-dimensional (3D) dECM using gentle decellularization techniques. This study demonstrated a crucial advancement-the retention of endogenous GAGs-enabling direct growth factor incorporation without chemical agents. As a proof-of-concept, brain-derived neurotrophic factor (BDNF) was incorporated into the 3D dECM to enhance its therapeutic potential for brain repair. In vitro, BDNF-loaded 3D dECM enabled sustained growth factor release, significantly enhancing the proneuritogenic, neuroprotective, and proangiogenic effects. In a mouse model of traumatic brain injury, the implantation of BDNF-loaded 3D dECM significantly enhanced motor function and facilitated brain repair. These findings highlight the adaptability of MSC spheroid-derived 3D dECM for tissue-specific customization through straightforward and translatable growth factor incorporation, demonstrating its potential as a pro-regenerative biomaterial for advancing regenerative medicine applications.
Collapse
Affiliation(s)
- Ying-Chi Kao
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Pei-Ching Yang
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Yu-Ping Lin
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Grace H Chen
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Shao-Wen Liu
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Chia-Hsin Ho
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Shih-Chen Huang
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Peng-Ying Lee
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Linyi Chen
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, 30013, Taiwan; Department of Medical Science, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Chieh-Cheng Huang
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan.
| |
Collapse
|
2
|
Tavares DF, Mano JF, Oliveira MB. Advances in abiotic tissue-based biomaterials: A focus on decellularization and devitalization techniques. Mater Today Bio 2025; 32:101735. [PMID: 40275948 PMCID: PMC12020859 DOI: 10.1016/j.mtbio.2025.101735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/14/2025] [Accepted: 04/05/2025] [Indexed: 04/26/2025] Open
Abstract
This Review explores the growing and diversifying field of tissue-derived abiotic constructs for tissue engineering applications, with main focus on decellularization and devitalization techniques and principles. Acellular fractions derived from biological tissues, such as the extracellular matrix (ECM), have long been considered a valuable approach for the generation of numerous scaffolds and more complex constructs. The removal of the cellular content has been considered essential to prevent the development of adverse immunological reactions. Nevertheless, the discovery of promising features of certain cellular components has sparked interest in the use of inactivated or devitalized cellular fractions for several applications, particularly in regenerative medicine and inflammation control. Devitalization has been described for several clinical applications, but remains poorly explored in terms of in vitro constructs compared to decellularization methods currently available. In this review, we present and critically evaluate a spectrum of approaches for the decellularization of whole-organs and in vitro constructs, and the most prevalent devitalization techniques, with a discussion on their implications on scaffolds composition, structure, and potentially therapeutic properties. Processing methodologies to achieve optimal cell-based abiotic materials and approaches for their effective characterization are described and discussed. The application of these materials in healthcare, with most focus on regenerative approaches and including examples of commercially available products, is also addressed.
Collapse
Affiliation(s)
- Diana F. Tavares
- Department of Chemistry, CICECO – Aveiro Institute of Materials. University of Aveiro., Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - João F. Mano
- Department of Chemistry, CICECO – Aveiro Institute of Materials. University of Aveiro., Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - Mariana B. Oliveira
- Department of Chemistry, CICECO – Aveiro Institute of Materials. University of Aveiro., Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| |
Collapse
|
3
|
Ho CT, Kao YC, Shyu YM, Wang IC, Liu QX, Liu SW, Huang SC, Chiu H, Hsu LW, Hsu TS, Hsieh WC, Huang CC. Assembly of MSCs into a spheroid configuration increases poly(I:C)-mediated TLR3 activation and the immunomodulatory potential of MSCs for alleviating murine colitis. Stem Cell Res Ther 2025; 16:172. [PMID: 40221757 PMCID: PMC11993957 DOI: 10.1186/s13287-025-04297-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 03/31/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Inflammatory bowel disease (IBD) is associated with significant clinical challenges due to the limitations of current therapeutic approaches. Mesenchymal stem cell (MSC)-based therapies have shown promise in alleviating IBD owing to their potent immunomodulatory properties. However, the therapeutic efficacy of these cells remains suboptimal, primarily due to the harsh peritoneal microenvironment, which compromises MSC viability and functional capacity after transplantation. METHODS To address these limitations, this study aimed to improve MSC engraftment and functionality by assembling MSCs into three-dimensional (3D) spheroids and priming them with the Toll-like receptor 3 (TLR3) agonist polyinosinic-polycytidylic acid (poly(I:C)). Their potential for treating IBD was evaluated using male C57BL/6 mice with dextran sulfate sodium-induced colitis. RESULTS While 3D spheroid formation alone upregulated TLR3 expression and increased MSC survival under oxidative stress, poly(I:C) priming had a pronounced synergistic effect, significantly increasing MSC-mediated splenocyte modulation and oxidative stress resistance. In a murine colitis model, compared with unprimed spheroids or MSC suspensions, poly(I:C)-primed MSC spheroids administered intraperitoneally exhibited increased survival and therapeutic efficacy, effectively alleviating colitis symptoms, reducing colonic inflammation, and promoting tissue recovery. CONCLUSION Collectively, these findings highlight the synergistic benefits of combining 3D spheroid assembly with TLR3 activation as an innovative strategy to improve the therapeutic efficacy of MSC-based treatments for IBD and other inflammatory diseases by increasing post-engraftment cell survival and immunomodulatory capacity.
Collapse
Affiliation(s)
- Chao-Ting Ho
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Ying-Chi Kao
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Yueh-Ming Shyu
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - I-Ching Wang
- Institute of Molecular and Cellular Biology, College of Life Sciences and Medicine, National Tsing Hua University, Hsinchu, Taiwan
| | - Qiao-Xuan Liu
- Institute of Molecular and Cellular Biology, College of Life Sciences and Medicine, National Tsing Hua University, Hsinchu, Taiwan
| | - Shao-Wen Liu
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Shih-Chen Huang
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Han Chiu
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Li-Wen Hsu
- Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu , Taiwan
| | - Tzu-Sheng Hsu
- Institute of Molecular and Cellular Biology, College of Life Sciences and Medicine, National Tsing Hua University, Hsinchu, Taiwan.
| | - Wan-Chen Hsieh
- Institute of Molecular and Cellular Biology, College of Life Sciences and Medicine, National Tsing Hua University, Hsinchu, Taiwan.
| | - Chieh-Cheng Huang
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
4
|
Chen GH, Sia KC, Liu SW, Kao YC, Yang PC, Ho CH, Huang SC, Lee PY, Liang MZ, Chen L, Huang CC. Implantation of MSC spheroid-derived 3D decellularized ECM enriched with the MSC secretome ameliorates traumatic brain injury and promotes brain repair. Biomaterials 2025; 315:122941. [PMID: 39515193 DOI: 10.1016/j.biomaterials.2024.122941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 09/14/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
Traumatic brain injury (TBI) presents substantial clinical challenges, as existing treatments are unable to reverse damage or effectively promote brain tissue regeneration. Although implantable biomaterials have been proposed to support tissue repair by mitigating the adverse microenvironment in injured brains, many fail to replicate the complex composition and architecture of the native extracellular matrix (ECM), resulting in only limited therapeutic outcomes. This study introduces an innovative approach by developing a mesenchymal stem cell (MSC) spheroid-derived three-dimensional (3D) decellularized ECM (dECM) that is enriched with the MSC-derived matrisome and secretome, offering a promising solution for TBI treatment and brain tissue regeneration. Proteomic and cytokine array analyses revealed that 3D dECM retained a diverse array of MSC spheroid-derived matrisome proteins and secretome components, which are crucial for replicating the complexity of native ECM and the therapeutic capabilities of MSCs. These molecules were found to underlie the observed effects of 3D dECM on immunomodulation, proneuritogenesis, and proangiogenesis in our in vitro functional assays. Implantation of 3D dECM into TBI model mice effectively mitigated postinjury tissue damage and promoted brain repair, as evidenced by a reduced brain lesion volume, decreased cell apoptosis, alleviated neuroinflammation, reduced glial scar formation, and increased of neuroblast recruitment to the lesion site. These outcomes culminated in improved motor function recovery in animals, highlighting the multifaceted therapeutic potential of 3D dECM for TBI. In summary, our study elucidates the transformative potential of MSC spheroid-derived bioactive 3D dECM as an implantable biomaterial for effectively mitigating post-TBI neurological damage, paving the way for its broader therapeutic application.
Collapse
Affiliation(s)
- Grace H Chen
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Kee-Chin Sia
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Shao-Wen Liu
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Ying-Chi Kao
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Pei-Ching Yang
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Chia-Hsin Ho
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Shih-Chen Huang
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Peng-Ying Lee
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Min-Zong Liang
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Linyi Chen
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, 30013, Taiwan; Department of Medical Science, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Chieh-Cheng Huang
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan.
| |
Collapse
|
5
|
Huang CC, Chang CK, Yang PC, Chiu H, Chen SH, Hsu LW. Injectable Glucose-Releasing Microgels Enhance the Survival and Therapeutic Potential of Transplanted MSCs Under Ischemic Conditions. Adv Healthc Mater 2025; 14:e2401724. [PMID: 39324547 DOI: 10.1002/adhm.202401724] [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: 05/09/2024] [Revised: 09/06/2024] [Indexed: 09/27/2024]
Abstract
Mesenchymal stem cell (MSC)-based therapies show potential to treat ischemic diseases owing to their versatile functions. However, sustaining MSC viability and therapeutic efficacy in ischemic tissues postengraftment remains a significant challenge. This is because, although MSCs are metabolically flexible, they fail to adapt to hypoxic conditions in the absence of glucose, leading to cell death. To overcome these issues, it is aimed to establish an injectable glucose delivery system using starch and amyloglucosidase embedded in alginate microgels. Here, starch/amyloglucosidase (S/A) microgels are engineered to continuously release glucose for seven days via enzymatic hydrolysis, thereby supporting MSC functions under ischemic conditions. In vitro tests under oxygen/glucose-deprived conditions revealed that the S/A microgels not only maintained the viability and intracellular energy but also enhanced the pro-angiogenic and immunomodulatory functions of MSCs. In vivo data further confirmed the pro-survival and pro-angiogenic effects of S/A microgels on MSCs following subcutaneous engraftment in mice. Overall, the developed S/A microgel significantly enhanced the survival and therapeutic potential of MSCs via sustained glucose delivery, highlighting its potential use in advancing MSC-based therapies for ischemic conditions.
Collapse
Affiliation(s)
- Chieh-Cheng Huang
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - Chun-Kai Chang
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - Pei-Ching Yang
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - Han Chiu
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - Shih-Heng Chen
- Department of Plastic & Reconstructive Surgery, Linkou Chang Gung Memorial Hospital, Taoyuan, 333423, Taiwan
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan, 333323, Taiwan
| | - Li-Wen Hsu
- Bioresource Collection and Research Center, Food Industry Research and Development Institute, Hsinchu, 300193, Taiwan
| |
Collapse
|
6
|
Ramos-Rodriguez DH, Fok SW, Dorais CJ, Filler AC, Caserta M, Leach JK. Decellularized Extracellular Matrix Improves Mesenchymal Stromal Cell Spheroid Response to Chondrogenic Stimuli. Tissue Eng Part A 2025; 31:139-151. [PMID: 39556314 PMCID: PMC11971541 DOI: 10.1089/ten.tea.2024.0267] [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: 09/04/2024] [Accepted: 10/08/2024] [Indexed: 11/19/2024] Open
Abstract
Cartilage regeneration is hindered due to the low proliferative capacity of chondrocytes and the avascular nature of cartilaginous tissue. Mesenchymal stromal cells (MSCs) are widely studied for cartilage tissue engineering, and the aggregation of MSCs into high-density cell spheroids facilitates chondrogenic differentiation due to increased cell-cell contact. Despite the promise of MSCs, the field would benefit from improved strategies to regulate the chondrogenic potential of MSCs differentiated from induced pluripotent stem cells (iPSCs), which are advantageous for their capacity to yield large numbers of required cells. We previously demonstrated the ability of MSC-secreted extracellular matrix (ECM) to promote MSC chondrogenic differentiation, but the combinatorial effect of iPSC-derived MSC (iMSC) spheroids, iMSC-derived decellularized ECM (idECM), and other stimuli (e.g., oxygen tension and transforming growth factor [TGF]-β) on chondrogenic potential has not been described. Similar to MSCs, iMSCs secreted a collagen-rich ECM. When incorporated into spheroids, idECM increased spheroid diameter and promoted chondrogenic differentiation. The combination of idECM loading, chondrogenic media, and hypoxia enhanced glycosaminoglycan (GAG) content 1.6-fold (40.9 ± 4.6 ng vs. 25.6 ± 3.3 ng, p < 0.05) in iMSC spheroids. Compared with active TGF-β1, the presentation of latent TGF-β1 resulted in greater GAG content (26.6 ± 1.8 ng vs. 41.9 ± 4.3 ng, p < 0.01). Finally, we demonstrated the capacity of individual spheroids to self-assemble into larger constructs and undergo both chondrogenic and hypertrophic differentiation when maintained in lineage-inducing media. These results highlight the potential of idECM to enhance the efficacy of chondrogenic stimuli for improved cartilage regeneration using human MSCs and iMSCs.
Collapse
Affiliation(s)
| | - Shierly W. Fok
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, California, USA
- Department of Biomedical Engineering, University of California Davis, Davis, California, USA
| | - Connor J. Dorais
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, California, USA
| | - Andrea C. Filler
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, California, USA
- Department of Biomedical Engineering, University of California Davis, Davis, California, USA
| | - Mason Caserta
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, California, USA
| | - J. Kent Leach
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, California, USA
- Department of Biomedical Engineering, University of California Davis, Davis, California, USA
| |
Collapse
|
7
|
Jiang T, Liu Q, Xu EC, He SY, Liu HY, Tian C, Zhang LF, Yang ZL. Fibroblasts/three-dimensional scaffolds complexes promote wound healing in rats with skin defects. Tissue Barriers 2025; 13:2334544. [PMID: 38544287 PMCID: PMC11875492 DOI: 10.1080/21688370.2024.2334544] [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: 10/19/2023] [Revised: 03/11/2024] [Accepted: 03/20/2024] [Indexed: 03/03/2025] Open
Abstract
We aim to construct a three-dimensional nano-skin scaffold material in vitro and study its promoting effect on wound healing in vivo. In this study, hybrid constructs of three-dimensional (3D) scaffolds were successfully fabricated by combination of type I collagen (COL-1) and polylactic-glycolic acid (PLGA). Fibroblasts and human umbilical cord mesenchymal stem cells (hUCMSCs) were used to implanted into 3D scaffolds and constructed into SD skin scaffolds in vitro. Finally, the fibroblasts/scaffolds complexes were inoculated on the surface of rat wound skin to study the promoting effect of the complex on wound healing. In our study, we successfully built a 3D scaffold, which had a certain porosity. Meanwhile, the content of COL-1 in the cell supernatant of fibroblast/scaffold complexes was increased. Furthermore, the expression of F-actin, CD105, integrin β, VEGF, and COL-1 was up-regulated in hUCMSC/scaffold complexes compared with the control group. In vivo, fibroblast/scaffold complexes promoted wound healing in rats. Our data suggested that the collagen Ⅳ and vimentin were elevated and collagen fibers were neatly arranged in the fibroblast/scaffold complex group was significantly higher than that in the scaffold group. Taken together, fibroblast/scaffold complexes were expected to be novel materials for treating skin defects.
Collapse
Affiliation(s)
- Ting Jiang
- Department of Burn and Plastic Surgery, Nanchong Central Hospital, The Second Clinical College of North Sichuan Medical College, Nanchong, China
| | - Qiang Liu
- Department of Burn and Plastic Surgery, Nanchong Central Hospital, The Second Clinical College of North Sichuan Medical College, Nanchong, China
| | - Er-Chang Xu
- Department of Burn and Plastic Surgery, Nanchong Central Hospital, The Second Clinical College of North Sichuan Medical College, Nanchong, China
| | - Si-Yu He
- Department of Burn and Plastic Surgery, Nanchong Central Hospital, The Second Clinical College of North Sichuan Medical College, Nanchong, China
| | - Hong-Yan Liu
- Department of Burn and Plastic Surgery, Nanchong Central Hospital, The Second Clinical College of North Sichuan Medical College, Nanchong, China
| | - Chao Tian
- Department of Burn and Plastic Surgery, Nanchong Central Hospital, The Second Clinical College of North Sichuan Medical College, Nanchong, China
| | - Lan-Fang Zhang
- Department of Burn and Plastic Surgery, Nanchong Central Hospital, The Second Clinical College of North Sichuan Medical College, Nanchong, China
| | - Ze-Long Yang
- Department of Orthopaedic Surgery, Nanchong Central Hospital, The Second Clinical College of North Sichuan Medical College, Nanchong, China
| |
Collapse
|
8
|
Di Nubila A, Doulgkeroglou MN, Gurdal M, Korntner SH, Zeugolis DI. In vitro and in vivo assessment of a non-animal sourced chitosan scaffold loaded with xeno-free umbilical cord mesenchymal stromal cells cultured under macromolecular crowding conditions. BIOMATERIALS AND BIOSYSTEMS 2024; 16:100102. [PMID: 40225717 PMCID: PMC11993840 DOI: 10.1016/j.bbiosy.2024.100102] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/06/2024] [Accepted: 10/08/2024] [Indexed: 04/15/2025] Open
Abstract
There is an increasing demand to not only accelerate the development of advanced therapy tissue engineered medicines, but to also eliminate xenogeneic materials from their development cycle. With these in mind, herein we first assessed the influence of carrageenan as macromolecular crowding agent to enhance and accelerate extracellular matrix deposition in xeno-free human umbilical cord mesenchymal stromal cell cultures and we developed and characterised a non-animal sourced chitosan scaffold. Following appropriate in vitro experimentation, a splinted nude mouse wound healing model was used to assess wound closure and scar size of non-treated control, non-animal sourced chitosan scaffold, non-animal sourced chitosan scaffold loaded with xeno-free human umbilical cord mesenchymal stromal cells and non-animal sourced chitosan scaffold loaded with xeno-free human umbilical cord mesenchymal stromal cells cultured under macromolecular crowding conditions groups. Across all three donors, carrageenan supplementation significantly increased collagen deposition at day 5, day 8 and day 11 without affecting cell morphology, viability, DNA concentration and metabolic activity. Through freeze drying, a non-animal sourced chitosan sponge was developed with appropriate structural and mechanical properties for wound healing applications. In vitro biological analysis made apparent that neither the scaffold nor macromolecular crowding negatively impacted xeno-free human umbilical cord mesenchymal stromal cell metabolic activity and proliferation. In vivo biological analysis revealed no significant differences between the groups in wound closure and scar size, raising question about the suitability of the model. In any case, this work sets the foundations for the development of completely xeno-free tissue engineered medicines.
Collapse
Affiliation(s)
- Alessia Di Nubila
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL) and Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland
| | - Meletios-Nikolaos Doulgkeroglou
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL) and Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland
| | - Mehmet Gurdal
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular & Biomedical Research and School of Mechanical & Materials Engineering, University College Dublin (UCD), Dublin, Ireland
| | - Stefanie H. Korntner
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL) and Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland
| | - Dimitrios I. Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL) and Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular & Biomedical Research and School of Mechanical & Materials Engineering, University College Dublin (UCD), Dublin, Ireland
| |
Collapse
|
9
|
Hsiao CH, Huang HL, Liu HL, Huang CC, Su JC, Chen YH, Lin YH. Combining ultrasound technology with targeted fucoidan/arginine-gelatin nanoparticles loaded with doxorubicin to enhance therapeutic efficacy and modulate bioeffects in drug-resistant triple-negative breast cancer. Int J Biol Macromol 2024; 283:137764. [PMID: 39557262 DOI: 10.1016/j.ijbiomac.2024.137764] [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: 06/06/2024] [Revised: 11/04/2024] [Accepted: 11/15/2024] [Indexed: 11/20/2024]
Abstract
Triple-negative breast cancer (TNBC) presents formidable challenges due to its aggressive nature and high recurrence rates, compounded by the involvement of epithelial-mesenchymal transition (EMT) in its progression and metastasis. Standard chemotherapy, which typically employs doxorubicin (DOX), remains a primary treatment approach. However, multidrug resistance (MDR) mechanisms, which include ATP-binding cassette transporters and EMT, contribute to treatment failures. Ultrasound has emerged as a promising modality among the various strategies explored to address MDR in TNBC. It serves as a diagnostic tool and holds therapeutic potential by inducing various biological effects depending on the exposure level. Targeted nanoparticles offer a means to enhance drug delivery efficiency. Our study aims to advance ultrasound technology combined with biocompatible nanoparticles using simplified preparation methods to improve treatment outcomes for drug-resistant TNBC. In particular, employing DOX-loaded fucoidan/arginine-gelatin nanoparticles facilitated the targeted delivery of chemotherapy drugs to tumors by effectively interacting with P-selectin, resulting in tumor growth inhibition. Furthermore, these nanoparticles mitigated MDR and EMT, particularly when combined with ultrasound treatment. This integrated approach of nanoparticle delivery with ultrasonography opens up a promising and innovative avenue for clinical cancer research.
Collapse
Affiliation(s)
- Chi-Huang Hsiao
- Division of Medical Oncology and Hematology, Department of Internal Medicine, Far Eastern Memorial Hospital, New Taipei 220216, Taiwan
| | - Hau-Lun Huang
- Department of Pharmacy, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Hao-Li Liu
- Department of Electrical Engineering, Department of Biomedical Engineering, National Taiwan University, Taipei 106216, Taiwan
| | - Chieh-Cheng Huang
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 300044, Taiwan
| | - Jung-Chen Su
- Department of Pharmacy, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Yi-Hsuan Chen
- Institute of Biopharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Yu-Hsin Lin
- Department of Pharmacy, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; Medical Device Innovation and Translation Center, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404327, Taiwan.
| |
Collapse
|
10
|
Yang MC, Chin IL, Fang H, Drack A, Nour S, Choi YS, O'Connor AJ, Greening DW, Kalionis B, Heath DE. Tailored environments for directed mesenchymal stromal cell proliferation and differentiation using decellularized extracellular matrices in conjunction with substrate modulus. Acta Biomater 2024; 187:110-122. [PMID: 39181177 DOI: 10.1016/j.actbio.2024.08.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 08/04/2024] [Accepted: 08/16/2024] [Indexed: 08/27/2024]
Abstract
Decellularised extracellular matrix (dECM) produced by mesenchymal stromal cells (MSCs) is a promising biomaterial for improving the ex vivo expansion of MSCs. The dECMs are often deposited on high modulus surfaces such as tissue culture plastic or glass, and subsequent differentiation assays often bias towards osteogenesis. We tested the hypothesis that dECM deposited on substrates of varying modulus will produce cell culture environments that are tailored to promote the proliferation and/or lineage-specific differentiation of MSCs. dECM was produced on type I collagen-functionalised polyacrylamide hydrogels with discrete moduli (∼4, 10, and 40 kPa) or in a linear gradient of modulus that spans the same range, and the substrates were used as culture surfaces for MSCs. Fluorescence spectroscopy and mass spectrometry characterization revealed structural compositional changes in the dECM as a function of substrate modulus. Softer substrates (4 kPa) with dECM supported the largest number of MSCs after 7 days (∼1.6-fold increase compared to glass). Additionally, osteogenic differentiation was greatest on high modulus substrates (40 kPa and glass) with dECM. Nuclear translocation of YAP1 was observed on all surfaces with a modulus of 10 kPa or greater and may be a driver for the increased osteogenesis on the high modulus surfaces. These data demonstrate that dECM technology can be integrated with environmental parameters such as substrate modulus to improve/tailor MSC proliferation and differentiation during ex vivo culture. These results have potential impact in the improved expansion of MSCs for tailored therapeutic applications and in the development of advanced tissue engineering scaffolds. STATEMENT OF SIGNIFICANCE: Mesenchymal stromal cells (MSCs) are extensively used in tissue engineering and regenerative medicine due to their ability to proliferate, differentiate, and modulate the immune environment. Controlling MSC behavior is critical for advances in the field. Decellularised extracellular matrix (dECM) can maintain MSC properties in culture, increase their proliferation rate and capacity, and enhance their stimulated differentiation. Substrate stiffness is another key driver of cell function, and previous reports have primarily looked at dECM deposition and function on stiff substrates such as glass. Herein, we produce dECM on substrates of varying stiffness to create tailored environments that enhance desired MSC properties such as proliferation and differentiation. Additionally, we complete mechanistic studies including quantitative mass spec of the ECM to understand the biological function.
Collapse
Affiliation(s)
- Michael C Yang
- Department of Biomedical Engineering, Graeme Clark Institute, University of Melbourne, Parkville, VIC, Australia; Department of Maternal-Fetal Medicine Pregnancy Research Centre, Royal Women's Hospital, Parkville, VIC, Australia
| | - Ian L Chin
- School of Human Sciences, University of Western Australia, Crawley, Western Australia, Australia
| | - Haoyun Fang
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia; Department of Cardiometabolic Health, University of Melbourne, Parkville, VIC Australia
| | - Auriane Drack
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia; Department of Cardiometabolic Health, University of Melbourne, Parkville, VIC Australia
| | - Shirin Nour
- Department of Biomedical Engineering, Graeme Clark Institute, University of Melbourne, Parkville, VIC, Australia; Department of Chemical Engineering, Polymer Science Group, University of Melbourne, Parkville, VIC, Australia
| | - Yu Suk Choi
- School of Human Sciences, University of Western Australia, Crawley, Western Australia, Australia
| | - Andrea J O'Connor
- Department of Biomedical Engineering, Graeme Clark Institute, University of Melbourne, Parkville, VIC, Australia
| | - David W Greening
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia; Department of Cardiovascular Research, Translation, and Implementation, La Trobe University, Bundoora, VIC, Australia; Central Clinical School, Monash University, Clayton, VIC, Australia; Department of Cardiometabolic Health, University of Melbourne, Parkville, VIC Australia
| | - Bill Kalionis
- Department of Maternal-Fetal Medicine Pregnancy Research Centre, Royal Women's Hospital, Parkville, VIC, Australia; Department of Obstetrics, Gynaecology and Newborn Health, University of Melbourne, Parkville, VIC, Australia.
| | - Daniel E Heath
- Department of Biomedical Engineering, Graeme Clark Institute, University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
11
|
Rampin A, Rossoni A, Chaniotaki L, Gkiatas IS, Tzora A, Skoufos I, Diakakis N, Prassinos N, Zeugolis DI. Xenogeneic versus allogeneic serum and macromolecular crowding in human tenocyte cultures. Eur J Cell Biol 2024; 103:151445. [PMID: 39024989 DOI: 10.1016/j.ejcb.2024.151445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 07/09/2024] [Accepted: 07/12/2024] [Indexed: 07/20/2024] Open
Abstract
Allogeneic serum and tissue-specific extracellular matrix have been shown to maintain permanently differentiated cell phenotype in culture. This is of particular importance for human tenocytes, a cell population that readily loses its function during ex vivo culture. With these in mind, herein we extracted human tenocytes using either foetal bovine serum or human serum, cultured them in the absence and presence of carrageenan and Ficoll®, the most widely used macromolecular crowding agents (to induce tissue-specific extracellular matrix deposition), and assessed cellular function, via metabolic activity, viability, proliferation and immunofluorescence for collagen related molecules, non-collagenous molecules and transmembrane molecules. At day 7, longest time point assessed, neither carrageenan nor Ficoll® significantly affected metabolic activity, viability and proliferation in either serum and human serum significantly increased metabolic activity and proliferation. At day 7, in the absence of macromolecular crowding, cells in human serum deposited significantly lower collagen type VI, biglycan, versican and tenomodulin than cells in foetal bovine serum. Interestingly, at day 7, in comparison to the no macromolecular crowding group, carrageenan in foetal bovine serum induced the highest effect, as judged by the highest number of significantly increased molecules (collagen type I, collagen type IV, collagen type V, collagen type VI, transforming growth factor β1, matrix metalloproteinase 14, lumican, versican, scleraxis and integrin α2β1). These data, although contradict previous observations where human serum outperformed foetal bovine serum, at the same time, support the use of foetal bovine serum in the development of cell-based medicines.
Collapse
Affiliation(s)
- Andrea Rampin
- Department of Agriculture, University of Ioannina, Arta, Greece; School of Veterinary Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece; Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular & Biomedical Research and School of Mechanical & Materials Engineering, University College Dublin (UCD), Dublin, Ireland
| | - Andrea Rossoni
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular & Biomedical Research and School of Mechanical & Materials Engineering, University College Dublin (UCD), Dublin, Ireland
| | - Lefki Chaniotaki
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular & Biomedical Research and School of Mechanical & Materials Engineering, University College Dublin (UCD), Dublin, Ireland
| | - Ioannis S Gkiatas
- Department of Orthopaedic Surgery, School of Medicine, University of Ioannina, Ioannina, Greece
| | - Athina Tzora
- Department of Agriculture, University of Ioannina, Arta, Greece
| | - Ioannis Skoufos
- Department of Agriculture, University of Ioannina, Arta, Greece
| | - Nikolaos Diakakis
- School of Veterinary Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Nikitas Prassinos
- School of Veterinary Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Dimitrios I Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular & Biomedical Research and School of Mechanical & Materials Engineering, University College Dublin (UCD), Dublin, Ireland.
| |
Collapse
|
12
|
Shen W, Mao Y, Ge X, Xu J, Hu J, Ao F, Wu S, Yan P. PLA tissue-engineered scaffolds loaded with sustained-release active substance chitosan nanoparticles: Modeling BSA-bFGF as the active substance. Int J Biol Macromol 2024; 274:133120. [PMID: 38876244 DOI: 10.1016/j.ijbiomac.2024.133120] [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: 10/06/2023] [Revised: 06/10/2024] [Accepted: 06/10/2024] [Indexed: 06/16/2024]
Abstract
The utilization of basic fibroblast growth factor (bFGF) in the development of tissue-engineered scaffolds is both challenging and imperative. In our pursuit of creating a scaffold that aligns with the natural healing process, we initially fabricated chitosan-bFGF nanoparticles (CS-bFGF NPs) through electrostatic spraying. Subsequently, polylactic acid (PLA) fiber was prepared using electrospinning technique, and the CS-bFGF NPs were uniformly embedded within the pores of porous PLA fibers. Scanning electron micrographs illustrate the smooth surface of the nanoparticles, showing a porous structure intricately attached to PLA fibers. Fourier-transform infrared spectroscopy (FTIR), energy-dispersive X-ray spectroscopy (EDS), and X-ray diffraction (XRD) analyses provided conclusive evidence that the CS-bFGF NPs were uniformly distributed throughout the porous PLA fibers, forming a robust physical bond through electrostatic adsorption. The resultant scaffolds exhibited commendable mechanical properties and hydrophilicity, facilitating a sustained-release for 72 h. Furthermore, the biocompatibility and degradation performance of the scaffolds were substantiated by monitoring conductivity and pH changes in pure water over different time intervals, complemented by scanning electron microscopy (SEM) observations. Cell experiments confirmed the cytocompatibility of the scaffolds. In animal studies, the group treated with 16 % NPs/Scaffold demonstrated the highest epidermal reconstruction rate. In summary, our developed materials present a promising candidate for serving as a tissue engineering scaffold, showcasing exceptional biocompatibility, sustained-release characteristics, and substantial potential for promoting epidermal regeneration.
Collapse
Affiliation(s)
- Wen Shen
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science and Technology, Xi'an 710021, China.
| | - Yueyang Mao
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science and Technology, Xi'an 710021, China
| | - Xuemei Ge
- College of Light Industry and Food Engineering, Nanjing Forestry University Nanjing, Nan Jing 210037, China
| | - Jingwen Xu
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science and Technology, Xi'an 710021, China
| | - Jiaru Hu
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science and Technology, Xi'an 710021, China
| | - Fen Ao
- College of Bioresources Chemical and Materials Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China
| | - Shang Wu
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science and Technology, Xi'an 710021, China
| | - Pi Yan
- School of Biological and Pharmaceutical Sciences, Shaanxi University of Science and Technology, Xi'an 710021, China
| |
Collapse
|
13
|
Lit KK, Zhirenova Z, Blocki A. Insulin-like growth factor-binding protein 7 (IGFBP7): A microenvironment-dependent regulator of angiogenesis and vascular remodeling. Front Cell Dev Biol 2024; 12:1421438. [PMID: 39045455 PMCID: PMC11263173 DOI: 10.3389/fcell.2024.1421438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 06/10/2024] [Indexed: 07/25/2024] Open
Abstract
Insulin-like Growth Factor-Binding Protein 7 (IGFBP7) is an extracellular matrix (ECM) glycoprotein, highly enriched in activated vasculature during development, physiological and pathological tissue remodeling. Despite decades of research, its role in tissue (re-)vascularization is highly ambiguous, exhibiting pro- and anti-angiogenic properties in different tissue remodeling states. IGFBP7 has multiple binding partners, including structural ECM components, cytokines, chemokines, as well as several receptors. Based on current evidence, it is suggested that IGFBP7's bioactivity is strongly dependent on the microenvironment it is embedded in. Current studies indicate that during physiological angiogenesis, IGFBP7 promotes endothelial cell attachment, luminogenesis, vessel stabilization and maturation. Its effects on other stages of angiogenesis and vessel function remain to be determined. IGFBP7 also modulates the pro-angiogenic properties of other signaling factors, such as VEGF-A and IGF, and potentially acts as a growth factor reservoir, while its actual effects on the factors' signaling may depend on the environment IGFBP7 is embedded in. Besides (re-)vascularization, IGFBP7 clearly promotes progenitor and stem cell commitment and may exhibit anti-inflammatory and anti-fibrotic properties. Nonetheless, its role in inflammation, immunomodulation, fibrosis and cellular senescence is again likely to be context-dependent. Future studies are required to shed more light on the intricate functioning of IGFBP7.
Collapse
Affiliation(s)
- Kwok Keung Lit
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- Center for Neuromusculoskeletal Restorative Medicine (CNRM), Hong Kong Science Park, Shatin, Hong Kong SAR, China
| | - Zhamilya Zhirenova
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- Center for Neuromusculoskeletal Restorative Medicine (CNRM), Hong Kong Science Park, Shatin, Hong Kong SAR, China
| | - Anna Blocki
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- Center for Neuromusculoskeletal Restorative Medicine (CNRM), Hong Kong Science Park, Shatin, Hong Kong SAR, China
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| |
Collapse
|
14
|
Kang Y, Na J, Karima G, Amirthalingam S, Hwang NS, Kim HD. Mesenchymal Stem Cell Spheroids: A Promising Tool for Vascularized Tissue Regeneration. Tissue Eng Regen Med 2024; 21:673-693. [PMID: 38578424 PMCID: PMC11187036 DOI: 10.1007/s13770-024-00636-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 02/29/2024] [Accepted: 03/05/2024] [Indexed: 04/06/2024] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) are undifferentiated cells that can differentiate into specific cell lineages when exposed to the right conditions. The ability of MSCs to differentiate into particular cells is considered very important in biological research and clinical applications. MSC spheroids are clusters of MSCs cultured in three dimensions, which play an important role in enhancing the proliferation and differentiation of MSCs. MSCs can also participate in vascular formation by differentiating into endothelial cells and secreting paracrine factors. Vascularization ability is essential in impaired tissue repair and function recovery. Therefore, the vascularization ability of MSCs, which enhances angiogenesis and accelerates tissue healing has made MSCs a promising tool for tissue regeneration. However, MSC spheroids are a relatively new research field, and more research is needed to understand their full potential. METHODS In this review, we highlight the importance of MSC spheroids' vascularization ability in tissue engineering and regenerative medicine while providing the current status of studies on the MSC spheroids' vascularization and suggesting potential future research directions for MSC spheroids. RESULTS Studies both in vivo and in vitro have demonstrated MSC spheroids' capacity to develop into endothelial cells and stimulate vasculogenesis. CONCLUSION MSC spheroids show potential to enhance vascularization ability in tissue regeneration. Yet, further research is required to comprehensively understand the relationship between MSC spheroids and vascularization mechanisms.
Collapse
Affiliation(s)
- Yoonjoo Kang
- Department of IT Convergence (Brain Korea Plus 21), Korea National University of Transportation, Chungju, 27469, Republic of Korea
| | - Jinwoo Na
- Department of Polymer Science and Engineering, Korea National University of Transportation, 50 Daehak-ro, Chungju, 27469, Republic of Korea
| | - Gul Karima
- Department of Polymer Science and Engineering, Korea National University of Transportation, 50 Daehak-ro, Chungju, 27469, Republic of Korea
| | - Sivashanmugam Amirthalingam
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
- Institute of Engineering Research, Seoul National University, Seoul, 08826, Republic of Korea
| | - Nathaniel S Hwang
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
- Institute of Engineering Research, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hwan D Kim
- Department of IT Convergence (Brain Korea Plus 21), Korea National University of Transportation, Chungju, 27469, Republic of Korea.
- Department of Polymer Science and Engineering, Korea National University of Transportation, 50 Daehak-ro, Chungju, 27469, Republic of Korea.
- Department of Biomedical Engineering, Korea National University of Transportation, Chungju, 27469, Republic of Korea.
| |
Collapse
|
15
|
Rodríguez-Soto MA, Riveros-Cortés A, Orjuela-Garzón IC, Fernández-Calderón IM, Rodríguez CF, Vargas NS, Ostos C, Camargo CM, Cruz JC, Kim S, D’Amore A, Wagner WR, Briceño JC. Redefining vascular repair: revealing cellular responses on PEUU-gelatin electrospun vascular grafts for endothelialization and immune responses on in vitro models. Front Bioeng Biotechnol 2024; 12:1410863. [PMID: 38903186 PMCID: PMC11188488 DOI: 10.3389/fbioe.2024.1410863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 05/13/2024] [Indexed: 06/22/2024] Open
Abstract
Tissue-engineered vascular grafts (TEVGs) poised for regenerative applications are central to effective vascular repair, with their efficacy being significantly influenced by scaffold architecture and the strategic distribution of bioactive molecules either embedded within the scaffold or elicited from responsive tissues. Despite substantial advancements over recent decades, a thorough understanding of the critical cellular dynamics for clinical success remains to be fully elucidated. Graft failure, often ascribed to thrombogenesis, intimal hyperplasia, or calcification, is predominantly linked to improperly modulated inflammatory reactions. The orchestrated behavior of repopulating cells is crucial for both initial endothelialization and the subsequent differentiation of vascular wall stem cells into functional phenotypes. This necessitates the TEVG to provide an optimal milieu wherein immune cells can promote early angiogenesis and cell recruitment, all while averting persistent inflammation. In this study, we present an innovative TEVG designed to enhance cellular responses by integrating a physicochemical gradient through a multilayered structure utilizing synthetic (poly (ester urethane urea), PEUU) and natural polymers (Gelatin B), thereby modulating inflammatory reactions. The luminal surface is functionalized with a four-arm polyethylene glycol (P4A) to mitigate thrombogenesis, while the incorporation of adhesive peptides (RGD/SV) fosters the adhesion and maturation of functional endothelial cells. The resultant multilayered TEVG, with a diameter of 3.0 cm and a length of 11 cm, exhibits differential porosity along its layers and mechanical properties commensurate with those of native porcine carotid arteries. Analyses indicate high biocompatibility and low thrombogenicity while enabling luminal endothelialization and functional phenotypic behavior, thus limiting inflammation in in-vitro models. The vascular wall demonstrated low immunogenicity with an initial acute inflammatory phase, transitioning towards a pro-regenerative M2 macrophage-predominant phase. These findings underscore the potential of the designed TEVG in inducing favorable immunomodulatory and pro-regenerative environments, thus holding promise for future clinical applications in vascular tissue engineering.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Carlos Ostos
- Instituto de Química, Facultad de Ciencias Exactas y Naturales, Universidad de Antioquia, Medellín, Colombia
| | | | - Juan C. Cruz
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá, Colombia
| | - Seungil Kim
- McGowan Institute for Regenerative Medicine and Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
| | - Antonio D’Amore
- McGowan Institute for Regenerative Medicine and Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
| | - William R. Wagner
- McGowan Institute for Regenerative Medicine and Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
| | - Juan C. Briceño
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá, Colombia
- Department of Congenital Heart Disease and Cardiovascular Surgery, Fundación CardioInfantil Instituto de Cardiología, Bogotá, Colombia
| |
Collapse
|
16
|
Pardo A, Gomez‐Florit M, Davidson MD, Öztürk‐Öncel MÖ, Domingues RMA, Burdick JA, Gomes ME. Hierarchical Design of Tissue-Mimetic Fibrillar Hydrogel Scaffolds. Adv Healthc Mater 2024; 13:e2303167. [PMID: 38400658 PMCID: PMC11209813 DOI: 10.1002/adhm.202303167] [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: 09/20/2023] [Revised: 02/05/2024] [Indexed: 02/25/2024]
Abstract
Most tissues of the human body present hierarchical fibrillar extracellular matrices (ECMs) that have a strong influence over their physicochemical properties and biological behavior. Of great interest is the introduction of this fibrillar structure to hydrogels, particularly due to the water-rich composition, cytocompatibility, and tunable properties of this class of biomaterials. Here, the main bottom-up fabrication strategies for the design and production of hierarchical biomimetic fibrillar hydrogels and their most representative applications in the fields of tissue engineering and regenerative medicine are reviewed. For example, the controlled assembly/arrangement of peptides, polymeric micelles, cellulose nanoparticles (NPs), and magnetically responsive nanostructures, among others, into fibrillar hydrogels is discussed, as well as their potential use as fibrillar-like hydrogels (e.g., those from cellulose NPs) with key biofunctionalities such as electrical conductivity or remote stimulation. Finally, the major remaining barriers to the clinical translation of fibrillar hydrogels and potential future directions of research in this field are discussed.
Collapse
Affiliation(s)
- Alberto Pardo
- 3B's Research Group I3Bs – Research Institute on BiomaterialsBiodegradables and Biomimetics University of Minho Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine AvePark – Parque de Ciência e Tecnologia Zona Industrial da Gandra BarcoGuimarães4805‐017Portugal
- ICVS/3B's ‐ PT Government Associate LaboratoryBraga/Guimarães4710‐057Portugal
- Colloids and Polymers Physics GroupParticle Physics DepartmentMaterials Institute (iMATUS)and Health Research Institute (IDIS)University of Santiago de CompostelaSantiago de Compostela15782Spain
| | - Manuel Gomez‐Florit
- Health Research Institute of the Balearic Islands (IdISBa)Palma07010Spain
- Research Unit, Son Espases University Hospital (HUSE)Palma07010Spain
- Group of Cell Therapy and Tissue Engineering (TERCIT)Research Institute on Health Sciences (IUNICS)University of the Balearic Islands (UIB)Ctra. Valldemossa km 7.5Palma07122Spain
| | - Matthew D. Davidson
- BioFrontiers Institute and Department of Chemical and Biological EngineeringUniversity of Colorado BoulderBoulderCO80303USA
| | - Meftune Özgen Öztürk‐Öncel
- 3B's Research Group I3Bs – Research Institute on BiomaterialsBiodegradables and Biomimetics University of Minho Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine AvePark – Parque de Ciência e Tecnologia Zona Industrial da Gandra BarcoGuimarães4805‐017Portugal
- ICVS/3B's ‐ PT Government Associate LaboratoryBraga/Guimarães4710‐057Portugal
| | - Rui M. A. Domingues
- 3B's Research Group I3Bs – Research Institute on BiomaterialsBiodegradables and Biomimetics University of Minho Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine AvePark – Parque de Ciência e Tecnologia Zona Industrial da Gandra BarcoGuimarães4805‐017Portugal
- ICVS/3B's ‐ PT Government Associate LaboratoryBraga/Guimarães4710‐057Portugal
| | - Jason A. Burdick
- BioFrontiers Institute and Department of Chemical and Biological EngineeringUniversity of Colorado BoulderBoulderCO80303USA
| | - Manuela E. Gomes
- 3B's Research Group I3Bs – Research Institute on BiomaterialsBiodegradables and Biomimetics University of Minho Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine AvePark – Parque de Ciência e Tecnologia Zona Industrial da Gandra BarcoGuimarães4805‐017Portugal
- ICVS/3B's ‐ PT Government Associate LaboratoryBraga/Guimarães4710‐057Portugal
| |
Collapse
|
17
|
Fonseca VC, Van V, Ip BC. Primary Human Cell-Derived Extracellular Matrix from Decellularized Fibroblast Microtissues with Tissue-Dependent Composition and Microstructure. Cell Mol Bioeng 2024; 17:189-201. [PMID: 39050510 PMCID: PMC11263529 DOI: 10.1007/s12195-024-00809-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 06/18/2024] [Indexed: 07/27/2024] Open
Abstract
Purpose Human extracellular matrix (ECM) exhibits complex protein composition and architecture depending on tissue and disease state, which remains challenging to reverse engineer. One promising approach is based on cell-secreted ECM from primary human fibroblasts that can be decellularized into acellular biomaterials. However, fibroblasts cultured on rigid culture plastic or biomaterial scaffolds can experience aberrant mechanical cues that perturb the biochemical, mechanical, and the efficiency of ECM production. Methods Here, we demonstrate a method for preparing decellularized ECM using primary human fibroblasts with tissue and disease-specific features with two case studies: (1) cardiac fibroblasts; (2) lung fibroblasts from healthy or diseased donors. Cells aggregate into engineered microtissues in low adhesion microwells that deposited ECM and can be decellularized. We systematically investigate microtissue morphology, matrix architecture, and mechanical properties, along with transcriptomic and proteomic analysis. Results Microtissues exhibited tissue-specific gene expression and proteomics profiling, with ECM complexity similar to native tissues. Healthy lung microtissues exhibited web-like fibrillar collagen compared to dense patches in healthy heart microtissues. Diseased lung exhibited more disrupted collagen architecture than healthy. Decellularized microtissues had tissue-specific mechanical stiffness that was physiologically relevant. Importantly, decellularized microtissues supported viability and proliferation of human cells. Conclusions We show that engineered microtissues of primary human fibroblasts seeded in low-adhesion microwells can be decellularized to produce human, tissue and disease-specific ECM. This approach should be widely applicable for generating personalized matrix that recapitulate tissues and disease states, relevant for culturing patient cells ex vivo as well as implantation for therapeutic treatments. Supplementary Information The online version contains supplementary material available at 10.1007/s12195-024-00809-y.
Collapse
Affiliation(s)
- Vera C. Fonseca
- Department of Pathology & Laboratory Medicine, Brown University, Box G-E5, Providence, RI 02912 USA
| | - Vivian Van
- Department of Pathology & Laboratory Medicine, Brown University, Box G-E5, Providence, RI 02912 USA
| | - Blanche C. Ip
- Department of Pathology & Laboratory Medicine, Brown University, Box G-E5, Providence, RI 02912 USA
- Present Address: The Broad Institute of MIT and Harvard, 75 Ames Street Cambridge, Cambridge, MA 02142 USA
| |
Collapse
|
18
|
Xu Y, Yao Y, Gao J. Cell-Derived Matrix: Production, Decellularization, and Application of Wound Repair. Stem Cells Int 2024; 2024:7398473. [PMID: 38882595 PMCID: PMC11178417 DOI: 10.1155/2024/7398473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 04/25/2024] [Accepted: 05/17/2024] [Indexed: 06/18/2024] Open
Abstract
Chronic nonhealing wounds significantly reduce patients' quality of life and are a major burden on healthcare systems. Over the past few decades, tissue engineering materials have emerged as a viable option for wound healing, with cell-derived extracellular matrix (CDM) showing remarkable results. The CDM's compatibility and resemblance to the natural tissue microenvironment confer distinct advantages to tissue-engineered scaffolds in wound repair. This review summarizes the current processes for CDM preparation, various cell decellularization protocols, and common characterization methods. Furthermore, it discusses the applications of CDM in wound healing, including skin defect and wound repair, angiogenesis, and engineered vessels, and offers perspectives on future developments.
Collapse
Affiliation(s)
- Yidan Xu
- Department of Plastic and Cosmetic Surgery Nanfang Hospital Southern Medical University, 1838 Guangzhou North Road, Guangzhou 510515, Guangdong, China
| | - Yao Yao
- Department of Plastic and Cosmetic Surgery Nanfang Hospital Southern Medical University, 1838 Guangzhou North Road, Guangzhou 510515, Guangdong, China
| | - Jianhua Gao
- Department of Plastic and Cosmetic Surgery Nanfang Hospital Southern Medical University, 1838 Guangzhou North Road, Guangzhou 510515, Guangdong, China
| |
Collapse
|
19
|
Guillaumin S, Gurdal M, Zeugolis DI. Gums as Macromolecular Crowding Agents in Human Skin Fibroblast Cultures. Life (Basel) 2024; 14:435. [PMID: 38672707 PMCID: PMC11051389 DOI: 10.3390/life14040435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 03/10/2024] [Accepted: 03/19/2024] [Indexed: 04/28/2024] Open
Abstract
Even though tissue-engineered medicines are under intense academic, clinical, and commercial investigation, only a handful of products have been commercialised, primarily due to the costs associated with their prolonged manufacturing. While macromolecular crowding has been shown to enhance and accelerate extracellular matrix deposition in eukaryotic cell culture, possibly offering a solution in this procrastinating tissue-engineered medicine development, there is still no widely accepted macromolecular crowding agent. With these in mind, we herein assessed the potential of gum Arabic, gum gellan, gum karaya, and gum xanthan as macromolecular crowding agents in WS1 skin fibroblast cultures (no macromolecular crowding and carrageenan were used as a control). Dynamic light scattering analysis revealed that all macromolecules had negative charge and were polydispersed. None of the macromolecules affected basic cellular function. At day 7 (the longest time point assessed), gel electrophoresis analysis revealed that all macromolecules significantly increased collagen type I deposition in comparison to the non-macromolecular crowding group. Also at day 7, immunofluorescence analysis revealed that carrageenan; the 50 µg/mL, 75 µg/mL, and 100 µg/mL gum gellan; and the 500 µg/mL and 1000 µg/mL gum xanthan significantly increased both collagen type I and collagen type III deposition and only carrageenan significantly increased collagen type V deposition, all in comparison to the non-macromolecular crowding group at the respective time point. This preliminary study demonstrates the potential of gums as macromolecular crowding agents, but more detailed biological studies are needed to fully exploit their potential in the development of tissue-engineered medicines.
Collapse
Affiliation(s)
- Salome Guillaumin
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL) and Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, H91 TK33 Galway, Ireland; (S.G.); (M.G.)
| | - Mehmet Gurdal
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL) and Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, H91 TK33 Galway, Ireland; (S.G.); (M.G.)
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular & Biomedical Research and School of Mechanical & Materials Engineering, University College Dublin (UCD), D04 V1W8 Dublin, Ireland
| | - Dimitrios I. Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL) and Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, H91 TK33 Galway, Ireland; (S.G.); (M.G.)
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular & Biomedical Research and School of Mechanical & Materials Engineering, University College Dublin (UCD), D04 V1W8 Dublin, Ireland
| |
Collapse
|
20
|
Chen S, Wang H, Yang P, Chen S, Ho C, Yang P, Kao Y, Liu S, Chiu H, Lin Y, Chuang E, Huang J, Kao H, Huang C. Schwann cells acquire a repair phenotype after assembling into spheroids and show enhanced in vivo therapeutic potential for promoting peripheral nerve repair. Bioeng Transl Med 2024; 9:e10635. [PMID: 38435829 PMCID: PMC10905550 DOI: 10.1002/btm2.10635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 11/24/2023] [Accepted: 12/05/2023] [Indexed: 03/05/2024] Open
Abstract
The prognosis for postinjury peripheral nerve regeneration remains suboptimal. Although transplantation of exogenous Schwann cells (SCs) has been considered a promising treatment to promote nerve repair, this strategy has been hampered in practice by the limited availability of SC sources and an insufficient postengraftment cell retention rate. In this study, to address these challenges, SCs were aggregated into spheroids before being delivered to an injured rat sciatic nerve. We found that the three-dimensional aggregation of SCs induced their acquisition of a repair phenotype, as indicated by enhanced levels of c-Jun expression/activation and decreased expression of myelin sheath protein. Furthermore, our in vitro results demonstrated the superior potential of the SC spheroid-derived secretome in promoting neurite outgrowth of dorsal root ganglion neurons, enhancing the proliferation and migration of endogenous SCs, and recruiting macrophages. Moreover, transplantation of SC spheroids into rats after sciatic nerve transection effectively increased the postinjury nerve structure restoration and motor functional recovery rates, demonstrating the therapeutic potential of SC spheroids. In summary, transplantation of preassembled SC spheroids may hold great potential for enhancing the cell delivery efficiency and the resultant therapeutic outcome, thereby improving SC-based transplantation approaches for promoting peripheral nerve regeneration.
Collapse
Affiliation(s)
- Shih‐Heng Chen
- Department of Plastic and Reconstructive SurgeryLinkou Chang Gung Memorial HospitalTaoyuanTaiwan
- School of MedicineCollege of Medicine, Chang Gung UniversityTaoyuanTaiwan
| | - Hsin‐Wen Wang
- Institute of Biomedical EngineeringNational Tsing Hua UniversityHsinchuTaiwan
| | - Pei‐Ching Yang
- Department of Plastic and Reconstructive SurgeryLinkou Chang Gung Memorial HospitalTaoyuanTaiwan
| | - Shih‐Shien Chen
- Department of Plastic and Reconstructive SurgeryLinkou Chang Gung Memorial HospitalTaoyuanTaiwan
| | - Chia‐Hsin Ho
- Institute of Biomedical EngineeringNational Tsing Hua UniversityHsinchuTaiwan
| | - Pei‐Ching Yang
- Institute of Biomedical EngineeringNational Tsing Hua UniversityHsinchuTaiwan
| | - Ying‐Chi Kao
- Institute of Biomedical EngineeringNational Tsing Hua UniversityHsinchuTaiwan
| | - Shao‐Wen Liu
- Institute of Biomedical EngineeringNational Tsing Hua UniversityHsinchuTaiwan
| | - Han Chiu
- Institute of Biomedical EngineeringNational Tsing Hua UniversityHsinchuTaiwan
| | - Yu‐Jie Lin
- Institute of Biomedical EngineeringNational Tsing Hua UniversityHsinchuTaiwan
| | - Er‐Yuan Chuang
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, International Ph.D. Program in Biomedical Engineering, Taipei Medical UniversityTaipeiTaiwan
- Cell Physiology and Molecular Image Research CenterTaipei Medical University–Wan Fang HospitalTaipeiTaiwan
| | - Jen‐Huang Huang
- Department of Chemical EngineeringNational Tsing Hua UniversityHsinchuTaiwan
| | - Huang‐Kai Kao
- Department of Plastic and Reconstructive SurgeryLinkou Chang Gung Memorial HospitalTaoyuanTaiwan
- School of MedicineCollege of Medicine, Chang Gung UniversityTaoyuanTaiwan
| | - Chieh‐Cheng Huang
- Institute of Biomedical EngineeringNational Tsing Hua UniversityHsinchuTaiwan
| |
Collapse
|
21
|
Tevlek A. The role of decellularized cell derived extracellular matrix in the establishment and culture of in vitrobreast cancer tumor model. Biomed Mater 2024; 19:025037. [PMID: 38286003 DOI: 10.1088/1748-605x/ad2378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 01/29/2024] [Indexed: 01/31/2024]
Abstract
Decades of research have shown that two-dimensional cell culture studies are insufficient for preclinical cancer diagnosis and treatment, and that cancer cells in three-dimensional (3D) culture systems have better cell-cell and cell-matrix interactions, gene expression, heterogeneity, and structural complexity that more closely resemblein vivotumors. Researchers are still optimizing 3D culturing settings for different cancers. Despite promising tumor spheroid research, tumor cell-only aggregates lack the tumor microenvironment and cannot model tumors. Here, MCF-7 breast cancer cell derived decellularized extracellular matrix (CD-dECMs) were obtained and converted into autologous, biologically active, biocompatible, and non-immunogenic hydrogels to be used as micro-environment in both organoid formation and culture. For the production of organoids, CD-dECM doping concentrations ranging from 0.1 mg ml-1to 1.5 mg ml-1were evaluated, and the lowest concentration was found to be the most effective. For organoid culture, 8 mg ml-1CD-dECM, 4 mg ml-1rat tendon collagen type I (Col I) (4 mg ml-1) and a 1:1 (v/v) mixture of these two were used and the most viable and the biggest organoids were discovered in CD-dECM/Col I (1:1) group. The results show that autologous CD-dECM can replace hydrogels in tumor organoid generation and culture at low and high concentrations, respectively.
Collapse
Affiliation(s)
- Atakan Tevlek
- Middle East Technical University (METU), MEMS Research and Application Center, 06530 Ankara, Turkey
| |
Collapse
|
22
|
Chen SH, Lee YW, Kao HK, Yang PC, Chen SH, Liu SW, Yang PC, Lin YJ, Huang CC. The Transplantation of 3-Dimensional Spheroids of Adipose-Derived Stem Cells Promotes Achilles Tendon Healing in Rabbits by Enhancing the Proliferation of Tenocytes and Suppressing M1 Macrophages. Am J Sports Med 2024; 52:406-422. [PMID: 38193194 DOI: 10.1177/03635465231214698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2024]
Abstract
BACKGROUND Tendons have limited regenerative potential, so healing of ruptured tendon tissue requires a prolonged period, and the prognosis is suboptimal. Although stem cell transplantation-based approaches show promise for accelerating tendon repair, the resultant therapeutic efficacy remains unsatisfactory. HYPOTHESIS The transplantation of stem cells preassembled as 3-dimensional spheroids achieves a superior therapeutic outcome compared with the transplantation of single-cell suspensions. STUDY DESIGN Controlled laboratory study. METHODS Adipose-derived stem cells (ADSCs) were assembled as spheroids using a methylcellulose hydrogel system. The secretome of ADSC suspensions or spheroids was collected and utilized to treat tenocytes and macrophages to evaluate their therapeutic potential and investigate the mechanisms underlying their effects. RNA sequencing was performed to investigate the global difference in gene expression between ADSC suspensions and spheroids in an in vitro inflammatory microenvironment. For the in vivo experiment, rabbits that underwent Achilles tendon transection, followed by stump suturing, were randomly assigned to 1 of 3 groups: intratendinous injection of saline, rabbit ADSCs as conventional single-cell suspensions, or preassembled ADSC spheroids. The tendons were harvested for biomechanical testing and histological analysis at 4 weeks postoperatively. RESULTS Our in vitro results demonstrated that the secretome of ADSCs assembled as spheroids exhibited enhanced modulatory activity in (1) tenocyte proliferation (P = .015) and migration (P = .001) by activating extracellular signal-regulated kinase (ERK) signaling and (2) the suppression of the secretion of interleukin-6 (P = .005) and interleukin-1α (P = .042) by M1 macrophages via the COX-2/PGE2/EP4 signaling axis. Gene expression profiling of cells exposed to an inflammatory milieu revealed significantly enriched terms that were associated with the immune response, cytokines, and tissue remodeling in preassembled ADSC spheroids. Ex vivo fluorescence imaging revealed that the engraftment efficiency of ADSCs in the form of spheroids was higher than that of ADSCs in single-cell suspensions (P = .003). Furthermore, the transplantation of ADSC spheroids showed superior therapeutic effects in promoting the healing of sutured stumps, as evidenced by improvements in the tensile strength (P = .019) and fiber alignment (P < .001) of the repaired tendons. CONCLUSION The assembly of ADSCs as spheroids significantly advanced their potential to harness tenocytes and macrophages. As a proof of concept, this study clearly demonstrates the effectiveness of using ADSC spheroids to promote tendon regeneration. CLINICAL RELEVANCE The present study lays a foundation for future clinical applications of stem cell spheroid-based therapy for the management of tendon injuries.
Collapse
Affiliation(s)
- Shih-Heng Chen
- Department of Plastic and Reconstructive Surgery, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yun-Wei Lee
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Huang-Kai Kao
- Department of Plastic and Reconstructive Surgery, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Pei-Ching Yang
- Department of Plastic and Reconstructive Surgery, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Shih-Hsien Chen
- Department of Plastic and Reconstructive Surgery, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Shao-Wen Liu
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Pei-Ching Yang
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Yu-Jie Lin
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Chieh-Cheng Huang
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|
23
|
Luo P, Huang R, Wu Y, Liu X, Shan Z, Gong L, Deng S, Liu H, Fang J, Wu S, Wu X, Liu Q, Chen Z, Yeung KW, Qiao W, Chen S, Chen Z. Tailoring the multiscale mechanics of tunable decellularized extracellular matrix (dECM) for wound healing through immunomodulation. Bioact Mater 2023; 28:95-111. [PMID: 37250862 PMCID: PMC10209339 DOI: 10.1016/j.bioactmat.2023.05.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 05/10/2023] [Accepted: 05/11/2023] [Indexed: 05/31/2023] Open
Abstract
With the discovery of the pivotal role of macrophages in tissue regeneration through shaping the tissue immune microenvironment, various immunomodulatory strategies have been proposed to modify traditional biomaterials. Decellularized extracellular matrix (dECM) has been extensively used in the clinical treatment of tissue injury due to its favorable biocompatibility and similarity to the native tissue environment. However, most reported decellularization protocols may cause damage to the native structure of dECM, which undermines its inherent advantages and potential clinical applications. Here, we introduce a mechanically tunable dECM prepared by optimizing the freeze-thaw cycles. We demonstrated that the alteration in micromechanical properties of dECM resulting from the cyclic freeze-thaw process contributes to distinct macrophage-mediated host immune responses to the materials, which are recently recognized to play a pivotal role in determining the outcome of tissue regeneration. Our sequencing data further revealed that the immunomodulatory effect of dECM was induced via the mechnotrasduction pathways in macrophages. Next, we tested the dECM in a rat skin injury model and found an enhanced micromechanical property of dECM achieved with three freeze-thaw cycles significantly promoted the M2 polarization of macrophages, leading to superior wound healing. These findings suggest that the immunomodulatory property of dECM can be efficiently manipulated by tailoring its inherent micromechanical properties during the decellularization process. Therefore, our mechanics-immunomodulation-based strategy provides new insights into the development of advanced biomaterials for wound healing.
Collapse
Affiliation(s)
- Pu Luo
- Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - Ruoxuan Huang
- Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - You Wu
- Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - Xingchen Liu
- Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - Zhengjie Shan
- Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - Li Gong
- Instrumental Analysis Research Center, Sun Yat-sen University, Guangzhou, 510275, China
| | - Shudan Deng
- Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - Haiwen Liu
- Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - Jinghan Fang
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region
- Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma, The University of Hong Kong-Shenzhen Hospital, Shenzhen, 518058, China
| | - Shiyu Wu
- Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - Xiayi Wu
- Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - Quan Liu
- Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - Zetao Chen
- Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - Kelvin W.K. Yeung
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region
- Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma, The University of Hong Kong-Shenzhen Hospital, Shenzhen, 518058, China
| | - Wei Qiao
- Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Shoucheng Chen
- Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - Zhuofan Chen
- Hospital of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| |
Collapse
|
24
|
Fonseca VC, Van V, Ip BC. Primary Human Cell-Derived Extracellular Matrix from Decellularized Fibroblast Microtissues with Tissue-Dependent Composition and Microstructure. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.15.553420. [PMID: 37645710 PMCID: PMC10462104 DOI: 10.1101/2023.08.15.553420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Human extracellular matrix (ECM) exhibits complex protein composition and architecture depending on tissue and disease state, which remains challenging to reverse engineer. One promising approach is based on cell-secreted ECM from human fibroblasts, which can then be decellularized into an acellular biomaterial. However, fibroblasts initially seeded on rigid tissue culture plastic or biomaterial scaffolds experience aberrant mechanical cues that influence ECM deposition. Here, we show that engineered microtissues of primary human fibroblasts seeded in low-adhesion microwells can be decellularized to produce human, tissue-specific ECM. We investigate: 1) cardiac fibroblasts, as well as 2) lung fibroblasts from healthy, idiopathic fibrosis and chronic obstructive pulmonary disease donors. We demonstrate optimized culture and decellularization conditions, then characterize gene expression and protein composition. We further characterize ECM microstructure and mechanical properties. We envision that this method could be utilized for biomanufacturing of patient and tissue-specific ECM for organoid drug screening as well as implantable scaffolds. Impact In this study, we demonstrate a method for preparing decellularized matrix using primary human fibroblasts with tissue and disease-specific features. We aggregate single cell dispersions into engineered tissues using low adhesion microwells and show culture conditions that promote ECM deposition. We demonstrate this approach for cardiac fibroblasts as well as lung fibroblasts (both normal and diseased). We systematically investigate tissue morphology, matrix architecture, and mechanical properties, along with transcriptomic and proteomic analysis. This approach should be widely applicable for generating personalized ECM with features of patient tissues and disease state, relevant for culturing patient cells ex vivo as well as implantation for therapeutic treatments.
Collapse
|
25
|
Kusova AM, Rakipov IT, Zuev YF. Effects of Homogeneous and Heterogeneous Crowding on Translational Diffusion of Rigid Bovine Serum Albumin and Disordered Alfa-Casein. Int J Mol Sci 2023; 24:11148. [PMID: 37446325 DOI: 10.3390/ijms241311148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/30/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023] Open
Abstract
Intracellular environment includes proteins, sugars, and nucleic acids interacting in restricted media. In the cytoplasm, the excluded volume effect takes up to 40% of the volume available for occupation by macromolecules. In this work, we tested several approaches modeling crowded solutions for protein diffusion. We experimentally showed how the protein diffusion deviates from conventional Brownian motion in artificial conditions modeling the alteration of medium viscosity and rigid spatial obstacles. The studied tracer proteins were globular bovine serum albumin and intrinsically disordered α-casein. Using the pulsed field gradient NMR, we investigated the translational diffusion of protein probes of different structures in homogeneous (glycerol) and heterogeneous (PEG 300/PEG 6000/PEG 40,000) solutions as a function of crowder concentration. Our results showed fundamentally different effects of homogeneous and heterogeneous crowded environments on protein self-diffusion. In addition, the applied "tracer on lattice" model showed that smaller crowding obstacles (PEG 300 and PEG 6000) create a dense net of restrictions noticeably hindering diffusing protein probes, whereas the large-sized PEG 40,000 creates a "less restricted" environment for the diffusive motion of protein molecules.
Collapse
Affiliation(s)
- Aleksandra M Kusova
- Kazan Institute of Biochemistry and Biophysics, FRC Kazan Scientific Center, Russian Academy of Sciences, Lobachevsky Str. 2/31, Kazan 420111, Russia
| | - Ilnaz T Rakipov
- Institute of Chemistry, Kazan Federal University, Kremlevskaya Str. 18, Kazan 420008, Russia
| | - Yuriy F Zuev
- Kazan Institute of Biochemistry and Biophysics, FRC Kazan Scientific Center, Russian Academy of Sciences, Lobachevsky Str. 2/31, Kazan 420111, Russia
- Institute of Chemistry, Kazan Federal University, Kremlevskaya Str. 18, Kazan 420008, Russia
| |
Collapse
|
26
|
Siddiqui AM, Thiele F, Stewart RN, Rangnick S, Weiss GJ, Chen BK, Silvernail JL, Strickland T, Nesbitt JJ, Lim K, Schwarzbauer JE, Schwartz J, Yaszemski MJ, Windebank AJ, Madigan NN. Open-Spaced Ridged Hydrogel Scaffolds Containing TiO 2-Self-Assembled Monolayer of Phosphonates Promote Regeneration and Recovery Following Spinal Cord Injury. Int J Mol Sci 2023; 24:10250. [PMID: 37373396 DOI: 10.3390/ijms241210250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/31/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
The spinal cord has a poor ability to regenerate after an injury, which may be due to cell loss, cyst formation, inflammation, and scarring. A promising approach to treating a spinal cord injury (SCI) is the use of biomaterials. We have developed a novel hydrogel scaffold fabricated from oligo(poly(ethylene glycol) fumarate) (OPF) as a 0.08 mm thick sheet containing polymer ridges and a cell-attractive surface on the other side. When the cells are cultured on OPF via chemical patterning, the cells attach, align, and deposit ECM along the direction of the pattern. Animals implanted with the rolled scaffold sheets had greater hindlimb recovery compared to that of the multichannel scaffold control, which is likely due to the greater number of axons growing across it. The immune cell number (microglia or hemopoietic cells: 50-120 cells/mm2 in all conditions), scarring (5-10% in all conditions), and ECM deposits (Laminin or Fibronectin: approximately 10-20% in all conditions) were equal in all conditions. Overall, the results suggest that the scaffold sheets promote axon outgrowth that can be guided across the scaffold, thereby promoting hindlimb recovery. This study provides a hydrogel scaffold construct that can be used in vitro for cell characterization or in vivo for future neuroprosthetics, devices, or cell and ECM delivery.
Collapse
Affiliation(s)
- Ahad M Siddiqui
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Frederic Thiele
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
- Program in Human Medicine, Paracelsus Medical Private University, 5020 Salzburg, Austria
| | - Rachel N Stewart
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
- Regenerative Medicine Institute (REMEDI), National University of Ireland Galway, H91 TK33 Galway, Ireland
| | - Simone Rangnick
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
- Program in Human Medicine, Paracelsus Medical Private University, 5020 Salzburg, Austria
| | - Georgina J Weiss
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
- Program in Human Medicine, Paracelsus Medical Private University, 90419 Nuremberg, Germany
| | - Bingkun K Chen
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Tammy Strickland
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
- Regenerative Medicine Institute (REMEDI), National University of Ireland Galway, H91 TK33 Galway, Ireland
| | | | - Kelly Lim
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
| | - Jean E Schwarzbauer
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Jeffrey Schwartz
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
| | | | | | | |
Collapse
|
27
|
Choi SH, Lee K, Han H, Mo H, Jung H, Ryu Y, Nam Y, Rim YA, Ju JH. Prochondrogenic effect of decellularized extracellular matrix secreted from human induced pluripotent stem cell-derived chondrocytes. Acta Biomater 2023:S1742-7061(23)00317-3. [PMID: 37295627 DOI: 10.1016/j.actbio.2023.05.052] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 05/18/2023] [Accepted: 05/30/2023] [Indexed: 06/12/2023]
Abstract
Cartilage is mainly composed of chondrocytes and the extracellular matrix (ECM), which exchange important biochemical and biomechanical signals necessary for differentiation and homeostasis. Human articular cartilage has a low ability for regeneration because it lacks blood vessels, nerves, and lymphatic vessels. Currently, cell therapeutics, including stem cells, provide a promising strategy for cartilage regeneration and treatment; however, there are various hurdles to overcome, such as immune rejection and teratoma formation. In this study, we assessed the applicability of the stem cell-derived chondrocyte ECM for cartilage regeneration. Human induced pluripotent stem cell (hiPSC)-derived chondrocytes (iChondrocytes) were differentiated, and decellularized ECM (dECM) was successfully isolated from cultured chondrocytes. Isolated dECM enhanced in vitro chondrogenesis of iPSCs when recellularized. Implanted dECM also restored osteochondral defects in a rat osteoarthritis model. A possible association with the glycogen synthase kinase-3 beta (GSK3β) pathway demonstrated the fate-determining importance of dECM in regulating cell differentiation. Collectively, we suggested the prochondrogenic effect of hiPSC-derived cartilage-like dECM and offered a promising approach as a non-cellular therapeutic for articular cartilage reconstruction without cell transplantation. STATEMENT OF SIGNIFICANCE: Human articular cartilage has low ability for regeneration and cell culture-based therapeutics could aid cartilage regeneration. Yet, the applicability of human induced pluripotent stem cell-derived chondrocyte (iChondrocyte) extracellular matrix (ECM) has not been elucidated. Therefore, we first differentiated iChondrocytes and isolated the secreted ECM by decellularization. Recellularization was performed to confirm the pro-chondrogenic effect of the decellularized ECM (dECM). In addition, we confirmed the possibility of cartilage repair by transplanting the dECM into the cartilage defect in osteochondral defect rat knee joint. We believe that our proof-of-concept study will serve as a basis for investigating the potential of dECM obtained from iPSC-derived differentiated cells as a non-cellular resource for tissue regeneration and other future applications.
Collapse
Affiliation(s)
- Si Hwa Choi
- Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea
| | | | - Heeju Han
- Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea
| | - Hyunkyung Mo
- Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea
| | | | - YoungWoo Ryu
- Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea
| | | | - Yeri Alice Rim
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea.
| | - Ji Hyeon Ju
- Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea; YiPSCELL, Inc., Seoul, South Korea; Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| |
Collapse
|
28
|
Wan HY, Chen JCH, Xiao Q, Wong CW, Yang B, Cao B, Tuan RS, Nilsson SK, Ho YP, Raghunath M, Kamm RD, Blocki A. Stabilization and improved functionality of three-dimensional perfusable microvascular networks in microfluidic devices under macromolecular crowding. Biomater Res 2023; 27:32. [PMID: 37076899 PMCID: PMC10116810 DOI: 10.1186/s40824-023-00375-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 04/04/2023] [Indexed: 04/21/2023] Open
Abstract
BACKGROUND There is great interest to engineer in vitro models that allow the study of complex biological processes of the microvasculature with high spatiotemporal resolution. Microfluidic systems are currently used to engineer microvasculature in vitro, which consists of perfusable microvascular networks (MVNs). These are formed through spontaneous vasculogenesis and exhibit the closest resemblance to physiological microvasculature. Unfortunately, under standard culture conditions and in the absence of co-culture with auxiliary cells as well as protease inhibitors, pure MVNs suffer from a short-lived stability. METHODS Herein, we introduce a strategy for stabilization of MVNs through macromolecular crowding (MMC) based on a previously established mixture of Ficoll macromolecules. The biophysical principle of MMC is based on macromolecules occupying space, thus increasing the effective concentration of other components and thereby accelerating various biological processes, such as extracellular matrix deposition. We thus hypothesized that MMC will promote the accumulation of vascular ECM (basement membrane) components and lead to a stabilization of MVN with improved functionality. RESULTS MMC promoted the enrichment of cellular junctions and basement membrane components, while reducing cellular contractility. The resulting advantageous balance of adhesive forces over cellular tension resulted in a significant stabilization of MVNs over time, as well as improved vascular barrier function, closely resembling that of in vivo microvasculature. CONCLUSION Application of MMC to MVNs in microfluidic devices provides a reliable, flexible and versatile approach to stabilize engineered microvessels under simulated physiological conditions.
Collapse
Affiliation(s)
- Ho-Ying Wan
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jack Chun Hin Chen
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Qinru Xiao
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Christy Wingtung Wong
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Boguang Yang
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Benjamin Cao
- Biomedical Manufacturing Commonwealth Scientific and Industrial Research Organisation (CSIRO), Melbourne, Australia
- Australian Regenerative Medicine Institute, Monash University, Melbourne, Australia
| | - Rocky S Tuan
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Center for Neuromusculoskeletal Restorative Medicine (CNRM), Hong Kong Science Park, Shatin, New Territories, Hong Kong SAR, China
| | - Susan K Nilsson
- Biomedical Manufacturing Commonwealth Scientific and Industrial Research Organisation (CSIRO), Melbourne, Australia
- Australian Regenerative Medicine Institute, Monash University, Melbourne, Australia
| | - Yi-Ping Ho
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Michael Raghunath
- Institute for Chemistry and Biotechnology, Zurich University of Applied Sciences, Wädenswil, Switzerland
| | - Roger D Kamm
- Department of Biology and Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Anna Blocki
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
- Center for Neuromusculoskeletal Restorative Medicine (CNRM), Hong Kong Science Park, Shatin, New Territories, Hong Kong SAR, China.
| |
Collapse
|
29
|
Zhu H, Wu X, Liu R, Zhao Y, Sun L. ECM-Inspired Hydrogels with ADSCs Encapsulation for Rheumatoid Arthritis Treatment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206253. [PMID: 36683217 PMCID: PMC10037981 DOI: 10.1002/advs.202206253] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/27/2022] [Indexed: 06/17/2023]
Abstract
Due to their intrinsic anti-inflammatory and immunomodulatory properties, adipose-derived stem cells (ADSCs) are explored as a promising alternative in treating rheumatoid arthritis (RA). To address the poor survival and function loss of directly injected stem cells, efforts in this area are focus on the generation of efficient cell delivery vehicles. Herein, a novel extracellular matrix (ECM)-inspired injectable hydrogel for ADSCs encapsulation and RA treatment is proposed. The hydrogel with dendritic polylysine and polysaccharide components is formed through the reversible Schiff base crosslinking. It possesses self-healing capability, superior mechanical properties, minimal toxicity, and immunomodulatory ability. When encapsulated with ADSCs, the hydrogel could recover chronic inflammation by directly reversing the dominant macrophage phenotype from M1 to M2 and inhibiting the migration of fibroblast-like synoviocytes. Through a collagen-induced arthritis rat model, the tremendous therapeutic outcomes of this ADSCs-laden hydrogel, including inflammation attenuation, cartilage protection, and bone mineral density promotion are demonstrated. These results make the ECM-inspired hydrogel laden with ADSCs an ideal candidate for treating RA and other autoimmune disorders.
Collapse
Affiliation(s)
- Haofang Zhu
- Department of Rheumatology and ImmunologyInstitute of Translational MedicineThe Affiliated Drum Tower Hospital of Nanjing University Medical School321 Zhongshan RoadNanjing210008P. R. China
- Department of Rheumatology and ImmunologyThe First Affiliated Hospital of Anhui Medical University218 Jixi RoadHefei230022P. R. China
| | - Xiangyi Wu
- Department of Rheumatology and ImmunologyInstitute of Translational MedicineThe Affiliated Drum Tower Hospital of Nanjing University Medical School321 Zhongshan RoadNanjing210008P. R. China
| | - Rui Liu
- Department of Rheumatology and ImmunologyInstitute of Translational MedicineThe Affiliated Drum Tower Hospital of Nanjing University Medical School321 Zhongshan RoadNanjing210008P. R. China
| | - Yuanjin Zhao
- Department of Rheumatology and ImmunologyInstitute of Translational MedicineThe Affiliated Drum Tower Hospital of Nanjing University Medical School321 Zhongshan RoadNanjing210008P. R. China
- State Key Laboratory of BioelectronicsSchool of Biological Science and Medical EngineeringSoutheast University2 SipailouNanjing210096P. R. China
| | - Lingyun Sun
- Department of Rheumatology and ImmunologyInstitute of Translational MedicineThe Affiliated Drum Tower Hospital of Nanjing University Medical School321 Zhongshan RoadNanjing210008P. R. China
- Department of Rheumatology and ImmunologyThe First Affiliated Hospital of Anhui Medical University218 Jixi RoadHefei230022P. R. China
| |
Collapse
|
30
|
Zhang Y, Feng Y, Shao Q, Jiang Z, Yang G. Rapid formation of 3D: Decellularized extracellular matrix spheroids for enhancing bone formation. J Biomed Mater Res A 2023; 111:378-388. [PMID: 36355784 DOI: 10.1002/jbm.a.37471] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 08/29/2022] [Accepted: 10/31/2022] [Indexed: 11/12/2022]
Abstract
Bone marrow mesenchymal stem cell sheet-derived spheroids (BMSCs spheroids) have been widely studied as native bioactive scaffolds. However, the abundant cells in BMSCs spheroids cause immunogenicity and make them difficult to store. This paper aimed to construct a new bioactive scaffold called 3D-decellularized extracellular matrix spheroids (ECM spheroids) via decellularization of BMSCs spheroids to enhance bone formation. Hematoxylin and eosin staining (HE), nuclear and cytoskeletal fluorescence, immunofluorescence (IF), and scanning electron microscopy (SEM) were utilized to detect the characteristics and components of ECM spheroids. Furthermore, the biological properties of migration, adhesion, and recellularization of cells in ECM spheroids were assessed in vitro, and bone formation was evaluated in rat calvarial defects. The results showed that both the nuclei and cytoskeleton in ECM spheroids were greatly altered and one of the major components of FN was intact. The migration, adhesion, and recellularization potential were improved in vitro. Meanwhile, ECM spheroids promoted osteogenesis in rat skull defects after 3 months (p < .01). In conclusion, ECM spheroids were successfully prepared and proven to promote cell migration, adhesion, and proliferation. Bone formation in vivo was also accelerated. We believe that ECM spheroids can be used as bioactive and biocompatible 3D scaffolds in the future.
Collapse
Affiliation(s)
- Yanmin Zhang
- Department of Stomatology, Integrated Traditional and Western Medicine Hospital of Linping District, Hangzhou, China
| | - Yuting Feng
- Department of Preventive Dentistry, Affiliated Hospital of Stomatology, Medical College, Zhejiang University, Zhejiang, Hangzhou, China.,Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, China
| | - Qin Shao
- Department of Stomatology, Integrated Traditional and Western Medicine Hospital of Linping District, Hangzhou, China
| | - Zhiwei Jiang
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, China.,Department of Implantology, Affiliated Hospital of Stomatology, Medical College, Zhejiang University, Zhejiang, Hangzhou, China
| | - Guoli Yang
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, China.,Department of Implantology, Affiliated Hospital of Stomatology, Medical College, Zhejiang University, Zhejiang, Hangzhou, China
| |
Collapse
|
31
|
Dong H, Li Z, Bian S, Song G, Song W, Zhang M, Xie H, Zheng S, Yang X, Li T, Song P. Culture of patient-derived multicellular clusters in suspended hydrogel capsules for pre-clinical personalized drug screening. Bioact Mater 2022; 18:164-177. [PMID: 35387168 PMCID: PMC8961426 DOI: 10.1016/j.bioactmat.2022.03.020] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 03/10/2022] [Accepted: 03/13/2022] [Indexed: 12/12/2022] Open
Abstract
A personalized medication regimen provides precise treatment for an individual and can be guided by pre-clinical drug screening. The economical and high-efficiency simulation of the liver tumor microenvironment (TME) in a drug-screening model has high value yet challenging to accomplish. Herein, we propose a simulation of the liver TME with suspended alginate-gelatin hydrogel capsules encapsulating patient-derived liver tumor multicellular clusters, and the culture of patient-derived tumor organoids(PDTOs) for personalized pre-clinical drug screening. The hydrogel capsule offers a 3D matrix environment with mechanical and biological properties similar to those of the liver in vivo. As a result, 18 of the 28 patient-derived multicellular clusters were successfully cultured as PDTOs. These PDTOs, along with hepatocyte growth factor (HGF) of non-cellular components, preserve stromal cells, including cancer-associated fibroblasts (CAFs) and vascular endothelial cells (VECs). They also maintain stable expression of molecular markers and tumor heterogeneity similar to those of the original liver tumors. Drugs, including cabazitaxel, oxaliplatin, and sorafenib, were tested in PDTOs. The sensitivity of PDTOs to these drugs differs between individuals. The sensitivity of one PDTO to oxaliplatin was validated using magnetic resonance imaging (MRI) and biochemical tests after oxaliplatin clinical treatment of the corresponding patient. Therefore, this approach is promising for economical, accurate, and high-throughput drug screening for personalized treatment.
Collapse
Affiliation(s)
- Haijiang Dong
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang Province, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Zhejiang Province, Hangzhou, 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Zhejiang Province, Hangzhou, 310003, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou, 310003, China
| | - Zequn Li
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang Province, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Zhejiang Province, Hangzhou, 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Zhejiang Province, Hangzhou, 310003, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou, 310003, China
| | - Suchen Bian
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang Province, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Zhejiang Province, Hangzhou, 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Zhejiang Province, Hangzhou, 310003, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou, 310003, China
| | - Guangyuan Song
- Department of Hepatopancreatobiliary Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Zhejiang Province, Hangzhou, 310014, China
| | - Wenfeng Song
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang Province, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Zhejiang Province, Hangzhou, 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Zhejiang Province, Hangzhou, 310003, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou, 310003, China
| | - Mingqi Zhang
- Center for X-Mechanics, Department of Engineering Mechanics, Zhejiang University, Zhejiang Province, Hangzhou, 310007, China
- Key Laboratory of Soft Machines and Smart Devices of Zhejiang Province, Zhejiang Province, Hangzhou, 310007, China
| | - Haiyang Xie
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang Province, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Zhejiang Province, Hangzhou, 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Zhejiang Province, Hangzhou, 310003, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou, 310003, China
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang Province, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Zhejiang Province, Hangzhou, 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Zhejiang Province, Hangzhou, 310003, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou, 310003, China
| | - Xuxu Yang
- Center for X-Mechanics, Department of Engineering Mechanics, Zhejiang University, Zhejiang Province, Hangzhou, 310007, China
- Key Laboratory of Soft Machines and Smart Devices of Zhejiang Province, Zhejiang Province, Hangzhou, 310007, China
| | - Tiefeng Li
- Center for X-Mechanics, Department of Engineering Mechanics, Zhejiang University, Zhejiang Province, Hangzhou, 310007, China
- Key Laboratory of Soft Machines and Smart Devices of Zhejiang Province, Zhejiang Province, Hangzhou, 310007, China
| | - Penghong Song
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang Province, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Zhejiang Province, Hangzhou, 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Zhejiang Province, Hangzhou, 310003, China
- Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou, 310003, China
| |
Collapse
|
32
|
Wong CWT, Sawhney A, Wu Y, Mak YW, Tian XY, Chan HF, Blocki A. Sourcing of human peripheral blood-derived myeloid angiogenic cells under xeno-free conditions for the treatment of critical limb ischemia. Stem Cell Res Ther 2022; 13:419. [PMID: 35964057 PMCID: PMC9375284 DOI: 10.1186/s13287-022-03095-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 07/26/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Critical limb ischemia (CLI) is the most severe form of peripheral artery disease and exhibits a high risk of lower extremity amputations. As even the most promising experimental approaches based on mesenchymal stem cells (MSCs) demonstrated only moderate therapeutic effects, we hypothesized that other cell types with intrinsic roles in angiogenesis may exhibit a stronger therapeutic potential. We have previously established a protocol to source human peripheral blood-derived angiogenic cells (BDACs). These cells promoted revascularization and took perivascular location at sites of angiogenesis, thus resembling hematopoietic pericytes, which were only described in vivo so far. We thus hypothesized that BDACs might have a superior ability to promote revascularization and rescue the affected limb in CLI. METHODS As standard BDAC sourcing techniques involve the use of animal-derived serum, we sought to establish a xeno- and/or serum-free protocol. Next, BDACs or MSCs were injected intramuscularly following the ligation of the iliac artery in a murine model. Their ability to enhance revascularization, impair necrosis and modulate inflammatory processes in the affected limb was investigated. Lastly, the secretomes of both cell types were compared to find potential indications for the observed differences in angiogenic potential. RESULTS From the various commercial media tested, one xeno-free medium enabled the derivation of cells that resembled functional BDACs in comparable numbers. When applied to a murine model of CLI, both cell types enhanced limb reperfusion and reduced necrosis, with BDACs being twice as effective as MSCs. This was also reflected in histological evaluation, where BDAC-treated animals exhibited the least muscle tissue degeneration. The BDAC secretome was enriched in a larger number of proteins with pro-angiogenic and anti-inflammatory properties, suggesting that the combination of those factors may be responsible for the superior therapeutic effect. CONCLUSIONS Functional BDACs can be sourced under xeno-free conditions paving the way for their safe clinical application. Since BDACs are derived from an easily accessible and renewable tissue, can be sourced in clinically relevant numbers and time frame and exceeded traditional MSCs in their therapeutic potential, they may represent an advantageous cell type for the treatment of CLI and other ischemic diseases.
Collapse
Affiliation(s)
- Christy Wing Tung Wong
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China.,School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Apurva Sawhney
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China.,School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Yalan Wu
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Yi Wah Mak
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China.,School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Xiao Yu Tian
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Hon Fai Chan
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China.,School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China.,Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Anna Blocki
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China. .,School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China. .,Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China.
| |
Collapse
|
33
|
Adeva-Andany MM, Carneiro-Freire N. Biochemical composition of the glomerular extracellular matrix in patients with diabetic kidney disease. World J Diabetes 2022; 13:498-520. [PMID: 36051430 PMCID: PMC9329837 DOI: 10.4239/wjd.v13.i7.498] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/19/2022] [Accepted: 06/26/2022] [Indexed: 02/06/2023] Open
Abstract
In the glomeruli, mesangial cells produce mesangial matrix while podocytes wrap glomerular capillaries with cellular extensions named foot processes and tether the glomerular basement membrane (GBM). The turnover of the mature GBM and the ability of adult podocytes to repair injured GBM are unclear. The actin cytoskeleton is a major cytoplasmic component of podocyte foot processes and links the cell to the GBM. Predominant components of the normal glomerular extracellular matrix (ECM) include glycosaminoglycans, proteoglycans, laminins, fibronectin-1, and several types of collagen. In patients with diabetes, multiorgan composition of extracellular tissues is anomalous, including the kidney, so that the constitution and arrangement of glomerular ECM is profoundly altered. In patients with diabetic kidney disease (DKD), the global quantity of glomerular ECM is increased. The level of sulfated proteoglycans is reduced while hyaluronic acid is augmented, compared to control subjects. The concentration of mesangial fibronectin-1 varies depending on the stage of DKD. Mesangial type III collagen is abundant in patients with DKD, unlike normal kidneys. The amount of type V and type VI collagens is higher in DKD and increases with the progression of the disease. The GBM contains lower amount of type IV collagen in DKD compared to normal tissue. Further, genetic variants in the α3 chain of type IV collagen may modulate susceptibility to DKD and end-stage kidney disease. Human cellular models of glomerular cells, analyses of human glomerular proteome, and improved microscopy procedures have been developed to investigate the molecular composition and organization of the human glomerular ECM.
Collapse
|
34
|
Lin SJ, Huang CC. Strontium Peroxide-Loaded Composite Scaffolds Capable of Generating Oxygen and Modulating Behaviors of Osteoblasts and Osteoclasts. Int J Mol Sci 2022; 23:ijms23116322. [PMID: 35683001 PMCID: PMC9181728 DOI: 10.3390/ijms23116322] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 06/01/2022] [Accepted: 06/03/2022] [Indexed: 11/24/2022] Open
Abstract
The reconstruction of bone defects remains challenging. The utilization of bone autografts, although quite promising, is limited by several drawbacks, especially substantial donor site complications. Recently, strontium (Sr), a bioactive trace element with excellent osteoinductive, osteoconductive, and pro-angiogenic properties, has emerged as a potential therapeutic agent for bone repair. Herein, a strontium peroxide (SrO2)-loaded poly(lactic-co-glycolic acid) (PLGA)-gelatin scaffold system was developed as an implantable bone substitute. Gelatin sponges serve as porous osteoconductive scaffolds, while PLGA not only reinforces the mechanical strength of the gelatin but also controls the rate of water infiltration. The encapsulated SrO2 can release Sr2+ in a sustained manner upon exposure to water, thus effectively stimulating the proliferation of osteoblasts and suppressing the formation of osteoclasts. Moreover, SrO2 can generate hydrogen peroxide and subsequent oxygen molecules to increase local oxygen tension, an essential niche factor for osteogenesis. Collectively, the developed SrO2-loaded composite scaffold shows promise as a multifunctional bioactive bone graft for bone tissue engineering.
Collapse
|
35
|
Feng Q, Li D, Li Q, Cao X, Dong H. Microgel assembly: Fabrication, characteristics and application in tissue engineering and regenerative medicine. Bioact Mater 2022; 9:105-119. [PMID: 34820559 PMCID: PMC8586262 DOI: 10.1016/j.bioactmat.2021.07.020] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/30/2021] [Accepted: 07/17/2021] [Indexed: 12/15/2022] Open
Abstract
Microgel assembly, a macroscopic aggregate formed by bottom-up assembly of microgels, is now emerging as prospective biomaterials for applications in tissue engineering and regenerative medicine (TERM). This mini-review first summarizes the fabrication strategies available for microgel assembly, including chemical reaction, physical reaction, cell-cell interaction and external driving force, then highlights its unique characteristics, such as microporosity, injectability and heterogeneity, and finally itemizes its applications in the fields of cell culture, tissue regeneration and biofabrication, especially 3D printing. The problems to be addressed for further applications of microgel assembly are also discussed.
Collapse
Affiliation(s)
- Qi Feng
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou, China
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, China
| | - Dingguo Li
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou, China
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou, China
- Guangdong Province Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, China
| | - Qingtao Li
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Xiaodong Cao
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou, China
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, China
| | - Hua Dong
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou, China
- National Engineering Research Center for Tissue Restoration and Reconstruction (NERC-TRR), Guangzhou, China
- Guangdong Province Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, China
| |
Collapse
|
36
|
Tan YH, Helms HR, Nakayama KH. Decellularization Strategies for Regenerating Cardiac and Skeletal Muscle Tissues. Front Bioeng Biotechnol 2022; 10:831300. [PMID: 35295645 PMCID: PMC8918733 DOI: 10.3389/fbioe.2022.831300] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/28/2022] [Indexed: 12/24/2022] Open
Abstract
Cardiovascular disease is the leading cause of death worldwide and is associated with approximately 17.9 million deaths each year. Musculoskeletal conditions affect more than 1.71 billion people globally and are the leading cause of disability. These two areas represent a massive global health burden that is perpetuated by a lack of functionally restorative treatment options. The fields of regenerative medicine and tissue engineering offer great promise for the development of therapies to repair damaged or diseased tissues. Decellularized tissues and extracellular matrices are cornerstones of regenerative biomaterials and have been used clinically for decades and many have received FDA approval. In this review, we first discuss and compare methods used to produce decellularized tissues and ECMs from cardiac and skeletal muscle. We take a focused look at how different biophysical properties such as spatial topography, extracellular matrix composition, and mechanical characteristics influence cell behavior and function in the context of regenerative medicine. Lastly, we describe emerging research and forecast the future high impact applications of decellularized cardiac and skeletal muscle that will drive novel and effective regenerative therapies.
Collapse
Affiliation(s)
| | | | - Karina H. Nakayama
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
37
|
Louisthelmy R, Burke BM, Cornelison RC. Brain Cancer Cell-Derived Matrices and Effects on Astrocyte Migration. Cells Tissues Organs 2022; 212:21-31. [PMID: 35168244 PMCID: PMC9376193 DOI: 10.1159/000522609] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 02/11/2022] [Indexed: 11/19/2022] Open
Abstract
Cell-derived matrices are useful tools for studying the extracellular matrix (ECM) of different cell types and testing the effects on cell migration or wound repair. These matrices typically are generated using extended culture with ascorbic acid to boost ECM production. Applying this technique to cancer cell cultures could advance the study of cancer ECM and its effects on recruitment and training of the tumor microenvironment, but ascorbic acid is potently cytotoxic to cancer cells. Macromolecular crowding (MMC) agents can also be added to increase matrix deposition based on the excluded volume principle. We report the use of MMC alone as an effective strategy to generate brain cancer cell-derived matrices for downstream analyses and cell migration studies. We cultured the mouse glioblastoma cell line GL261 for 1 week in the presence of three previously reported MMC agents (carrageenan, Ficoll 70/400, and hyaluronic acid). We measured the resulting deposition of collagens and sulfated glycosaminoglycans using quantitative assays, as well as other matrix components by immunostaining. Both carrageenan and Ficoll promoted significantly more accumulation of total collagen content, sulfated glycosaminoglycan content, and fibronectin staining. Only Ficoll, however, also demonstrated a significant increase in collagen I staining. The results were more variable in 3D spheroid culture. We focused on Ficoll MMC matrices, which were isolated using the small molecule Raptinal to induce cancer cell apoptosis and matrix decellularization. The cancer cell-derived matrix promoted significantly faster migration of human astrocytes in a scratch wound assay, which may be explained by focal adhesion morphology and an increase in cellular metabolic activity. Ultimately, these data show MMC culture is a useful technique to generate cancer cell-derived matrices and study the effects on stromal cell migration related to wound repair.
Collapse
Affiliation(s)
- Rebecca Louisthelmy
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA 10002
| | - Brycen M Burke
- Department of Biochemistry and Molecular Biology, University of Massachusetts Amherst, Amherst, MA 10002
| | - R Chase Cornelison
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA 10002
- Department of Chemical Engineering, University of Massachusetts Amherst, Amherst, MA 10002
- Neuroscience and Behavior Graduate Program, University of Massachusetts Amherst, Amherst, MA 10002
| |
Collapse
|
38
|
Nellinger S, Mrsic I, Keller S, Heine S, Southan A, Bach M, Volz A, Chassé T, Kluger PJ. Cell‐derived and enzyme‐based decellularized extracellular matrix exhibit compositional and structural differences that are relevant for its use as a biomaterial. Biotechnol Bioeng 2022; 119:1142-1156. [DOI: 10.1002/bit.28047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 01/01/2022] [Accepted: 01/19/2022] [Indexed: 11/11/2022]
Affiliation(s)
- Svenja Nellinger
- Reutlingen Research Institute Alteburgstr. 150 72762 Reutlingen Germany
| | - Ivana Mrsic
- Institute of Physical and Theoretical Chemistry, University of Tuebingen Auf der Morgenstelle 18 72076 Tuebingen Germany
| | - Silke Keller
- 3R‐Center for In Vitro Models and Alternatives to Animal Testing, Eberhard Karls University Tübingen Österbergstr. 3 72074 Tübingen Germany
- Department for Microphysiological Systems Institute of Biomedical Engineering, Faculty of Medicine of the Eberhard Karls University Tübingen Österbergstr. 3 72074 Tübingen Germany
| | - Simon Heine
- Reutlingen Research Institute Alteburgstr. 150 72762 Reutlingen Germany
| | - Alexander Southan
- Institute of Interfacial Process Engineering and Plasma Technology, University of Stuttgart Nobelstr. 12 70569 Stuttgart Germany
| | - Monika Bach
- Core Facility Hohenheim, University of Hohenheim Emil‐Wolff‐Str. 12 70599 Stuttgart Germany
| | - Ann‐Cathrin Volz
- Reutlingen Research Institute Alteburgstr. 150 72762 Reutlingen Germany
| | - Thomas Chassé
- Institute of Physical and Theoretical Chemistry, University of Tuebingen Auf der Morgenstelle 18 72076 Tuebingen Germany
| | - Petra J. Kluger
- School of Applied Chemistry, Reutlingen University Alteburgstr. 150 72762 Reutlingen Germany
| |
Collapse
|
39
|
Xu Z, Cao J, Zhao Z, Qiao Y, Liu X, Zhong J, Wang B, Suo G. A functional extracellular matrix biomaterial enriched with VEGFA and bFGF as vehicle of human umbilical cord mesenchymal stem cells in skin wound healing. Biomed Mater 2021; 17. [PMID: 34749352 DOI: 10.1088/1748-605x/ac37b0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 11/08/2021] [Indexed: 11/12/2022]
Abstract
The construction of microvascular network is one of the greatest challenges for tissue engineering and cell therapy. Endothelial cells are essential for the construction of network of blood vessels. However, their application meets challenges in clinic due to the limited resource of autologous endothelium. Mesenchymal stem cells can effectively promote the angiogenesis in ischemic tissues for their abilities of endothelial differentiation and paracrine, and abundant sources. Extracellular matrix (ECM) has been widely used as an ideal biomaterial to mimic cellular microenvironment for tissue engineering due to its merits of neutrality, good biocompatibility, degradability, and controllability. In this study, a functional cell derived ECM biomaterial enriched with VEGFA and bFGF by expressing the collagen-binding domain fused factor genes in host cells was prepared. This material could induce endothelial differentiation of human umbilical cord mesenchymal stem cells (hUCMSCs) and promote angiogenesis, which may improve the healing effect of skin injury. Our research not only provides a functional ECM material to inducing angiogenesis by inducing endothelial differentiation of hUCMSCs, but also shed light on the ubiquitous approaches to endow ECM materials different functions by enriching different factors. This study will benefit tissue engineering and regenerative medicine researches.
Collapse
Affiliation(s)
- Zhongjuan Xu
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, Anhui, People's Republic of China.,CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, Jiangsu, People's Republic of China
| | - Junjun Cao
- Livingchip Lnc., Nanjing 211112, Jiangsu, People's Republic of China
| | - Zhe Zhao
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, Jiangsu, People's Republic of China
| | - Yong Qiao
- Livingchip Lnc., Nanjing 211112, Jiangsu, People's Republic of China
| | - Xingzhi Liu
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, Anhui, People's Republic of China.,CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, Jiangsu, People's Republic of China
| | - Junjie Zhong
- Department of Neurosurgery, Fudan University Huashan Hospital, National Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai 200041, People's Republic of China
| | - Bin Wang
- Center for Clinic Stem Cell Research, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu, People's Republic of China
| | - Guangli Suo
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, Jiangsu, People's Republic of China
| |
Collapse
|
40
|
Modulation of Inherent Niches in 3D Multicellular MSC Spheroids Reconfigures Metabolism and Enhances Therapeutic Potential. Cells 2021; 10:cells10102747. [PMID: 34685727 PMCID: PMC8534378 DOI: 10.3390/cells10102747] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/05/2021] [Accepted: 10/12/2021] [Indexed: 01/02/2023] Open
Abstract
Multicellular spheroids show three-dimensional (3D) organization with extensive cell–cell and cell–extracellular matrix interactions. Owing to their native tissue-mimicking characteristics, mesenchymal stem cell (MSC) spheroids are considered promising as implantable therapeutics for stem cell therapy. Herein, we aim to further enhance their therapeutic potential by tuning the cultivation parameters and thus the inherent niche of 3D MSC spheroids. Significantly increased expression of multiple pro-regenerative paracrine signaling molecules and immunomodulatory factors by MSCs was observed after optimizing the conditions for spheroid culture. Moreover, these alterations in cellular behaviors may be associated with not only the hypoxic niche developed in the spheroid core but also with the metabolic reconfiguration of MSCs. The present study provides efficient methods for manipulating the therapeutic capacity of 3D MSC spheroids, thus laying solid foundations for future development and clinical application of spheroid-based MSC therapy for regenerative medicine.
Collapse
|
41
|
Rubí-Sans G, Nyga A, Rebollo E, Pérez-Amodio S, Otero J, Navajas D, Mateos-Timoneda MA, Engel E. Development of Cell-Derived Matrices for Three-Dimensional In Vitro Cancer Cell Models. ACS APPLIED MATERIALS & INTERFACES 2021; 13:44108-44123. [PMID: 34494824 DOI: 10.1021/acsami.1c13630] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Most morphogenetic and pathological processes are driven by cells responding to the surrounding matrix, such as its composition, architecture, and mechanical properties. Despite increasing evidence for the role of extracellular matrix (ECM) in tissue and disease development, many in vitro substitutes still fail to effectively mimic the native microenvironment. We established a novel method to produce macroscale (>1 cm) mesenchymal cell-derived matrices (CDMs) aimed to mimic the fibrotic tumor microenvironment surrounding epithelial cancer cells. CDMs are produced by human adipose mesenchymal stem cells cultured in sacrificial 3D scaffold templates of fibronectin-coated poly-lactic acid microcarriers (MCs) in the presence of macromolecular crowders. We showed that decellularized CDMs closely mimic the fibrillar protein composition, architecture, and mechanical properties of human fibrotic ECM from cancer masses. CDMs had highly reproducible composition made of collagen types I and III and fibronectin ECM with tunable mechanical properties. Moreover, decellularized and MC-free CDMs were successfully repopulated with cancer cells throughout their 3D structure, and following chemotherapeutic treatment, cancer cells showed greater doxorubicin resistance compared to 3D culture in collagen hydrogels. Collectively, these results support the use of CDMs as a reproducible and tunable tool for developing 3D in vitro cancer models.
Collapse
Affiliation(s)
- Gerard Rubí-Sans
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid 28029, Spain
| | - Agata Nyga
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain
| | - Elena Rebollo
- Molecular Imaging Platform, Molecular Biology Institute of Barcelona (IBMB-CSIC), Barcelona 08028, Spain
| | - Soledad Pérez-Amodio
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid 28029, Spain
- IMEM-BRT group, Department of Materials Science, EEBE, Technical University of Catalonia (UPC), Barcelona 08019, Spain
| | - Jorge Otero
- Unitat Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona 08036, Spain
- CIBER de Enfermedades Respiratorias, Madrid 28029, Spain
| | - Daniel Navajas
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain
- Unitat Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona 08036, Spain
- CIBER de Enfermedades Respiratorias, Madrid 28029, Spain
| | - Miguel A Mateos-Timoneda
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya (UIC), Sant Cugat del Vallès (Barcelona) 08195, Spain
| | - Elisabeth Engel
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona 08028, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid 28029, Spain
- IMEM-BRT group, Department of Materials Science, EEBE, Technical University of Catalonia (UPC), Barcelona 08019, Spain
| |
Collapse
|
42
|
Chen YC, Chen YH, Chiu H, Ko YH, Wang RT, Wang WP, Chuang YJ, Huang CC, Lu TT. Cell-Penetrating Delivery of Nitric Oxide by Biocompatible Dinitrosyl Iron Complex and Its Dermato-Physiological Implications. Int J Mol Sci 2021; 22:ijms221810101. [PMID: 34576264 PMCID: PMC8469893 DOI: 10.3390/ijms221810101] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/13/2021] [Accepted: 09/16/2021] [Indexed: 12/13/2022] Open
Abstract
After the discovery of endogenous dinitrosyl iron complexes (DNICs) as a potential biological equivalent of nitric oxide (NO), bioinorganic engineering of [Fe(NO)2] unit has emerged to develop biomimetic DNICs [(NO)2Fe(L)2] as a chemical biology tool for controlled delivery of NO. For example, water-soluble DNIC [Fe2(μ-SCH2CH2OH)2(NO)4] (DNIC-1) was explored for oral delivery of NO to the brain and for the activation of hippocampal neurogenesis. However, the kinetics and mechanism for cellular uptake and intracellular release of NO, as well as the biocompatibility of synthetic DNICs, remain elusive. Prompted by the potential application of NO to dermato-physiological regulations, in this study, cellular uptake and intracellular delivery of DNIC [Fe2(μ-SCH2CH2COOH)2(NO)4] (DNIC-2) and its regulatory effect/biocompatibility toward epidermal cells were investigated. Upon the treatment of DNIC-2 to human fibroblast cells, cellular uptake of DNIC-2 followed by transformation into protein-bound DNICs occur to trigger the intracellular release of NO with a half-life of 1.8 ± 0.2 h. As opposed to the burst release of extracellular NO from diethylamine NONOate (DEANO), the cell-penetrating nature of DNIC-2 rationalizes its overwhelming efficacy for intracellular delivery of NO. Moreover, NO-delivery DNIC-2 can regulate cell proliferation, accelerate wound healing, and enhance the deposition of collagen in human fibroblast cells. Based on the in vitro and in vivo biocompatibility evaluation, biocompatible DNIC-2 holds the potential to be a novel active ingredient for skincare products.
Collapse
Affiliation(s)
- Yu-Chieh Chen
- Department of Medical Science & Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu 30013, Taiwan; (Y.-C.C.); (Y.-H.K.); (Y.-J.C.)
| | - Yi-Hong Chen
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan; (Y.-H.C.); (H.C.)
| | - Han Chiu
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan; (Y.-H.C.); (H.C.)
| | - Yi-Hsuan Ko
- Department of Medical Science & Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu 30013, Taiwan; (Y.-C.C.); (Y.-H.K.); (Y.-J.C.)
| | - Ruei-Ting Wang
- CHLITINA Research and Development Center, CHLITINA Holding Ltd., Taipei 10073, Taiwan; (R.-T.W.); (W.-P.W.)
| | - Wei-Ping Wang
- CHLITINA Research and Development Center, CHLITINA Holding Ltd., Taipei 10073, Taiwan; (R.-T.W.); (W.-P.W.)
| | - Yung-Jen Chuang
- Department of Medical Science & Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu 30013, Taiwan; (Y.-C.C.); (Y.-H.K.); (Y.-J.C.)
| | - Chieh-Cheng Huang
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan; (Y.-H.C.); (H.C.)
- Correspondence: (C.-C.H.); (T.-T.L.)
| | - Tsai-Te Lu
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan; (Y.-H.C.); (H.C.)
- Correspondence: (C.-C.H.); (T.-T.L.)
| |
Collapse
|