1
|
Jiu J, Liu H, Li D, Li X, Zhang J, Yan L, Fan Z, Li S, Du G, Li JJ, Wu A, Liu W, Du Y, Zhao B, Wang B. 3D Mechanical Response Stem Cell Complex Repairs Spinal Cord Injury by Promoting Neurogenesis and Regulating Tissue Homeostasis. Adv Healthc Mater 2025; 14:e2404925. [PMID: 39853962 DOI: 10.1002/adhm.202404925] [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: 12/08/2024] [Indexed: 01/26/2025]
Abstract
Spinal cord injury (SCI) leads to acute tissue damage that disrupts the microenvironmental homeostasis of the spinal cord, inhibiting cell survival and function, and thereby undermining treatment efficacy. Traditional stem cell therapies have limited success in SCI, due to the difficulties in maintaining cell survival and inducing sustained differentiation into neural lineages. A new solution may arise from controlling the fate of stem cells by creating an appropriate mechanical microenvironment. In this study, mechanical response stem cell complex (MRSCC) is created as an innovative therapeutic strategy for SCI, utilizing 3D bioprinting technology and gelatin microcarriers (GM) loaded with mesenchymal stem cells (MSCs). GM creates an optimal microenvironment for MSCs growth and paracrine activity. Meanwhile, 3D bioprinting allows accurate control of spatial pore architecture and mechanical characteristics of the cell construct to encourage neuroregeneration. The mechanical microenvironment created by MRSCC is found to activate the Piezo1 channel and prevent excessive nuclear translocation of YAP, thereby increasing neural-related gene expression in MSCs. Transplanting MRSCC in rats with spinal cord injuries boosts sensory and motor recovery, reduces inflammation, and stimulates the regeneration of neurons and glial cells. The MRSCC offers a new tissue engineering solution that can promote spinal cord repair.
Collapse
Affiliation(s)
- Jingwei Jiu
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Haifeng Liu
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Dijun Li
- Department of Orthopedics, Affiliated Renhe Hospital of China Three Gorges University, Yichang, 443000, China
| | - Xiaoke Li
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Jing Zhang
- Department of Emergency Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550001, China
| | - Lei Yan
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Zijuan Fan
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, 030001, China
| | - Songyan Li
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Guangyuan Du
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Jiao Jiao Li
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Aimin Wu
- Department of Orthopaedics, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Wei Liu
- Development of Research, Beijing Hua Niche Biotechnology Co., LTD, Beijing, 100084, China
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Bin Zhao
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Bin Wang
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| |
Collapse
|
2
|
Jaime-Rodríguez M, Del Prado-Audelo ML, Sosa-Hernández NA, Anaya-Trejo DP, Villarreal-Gómez LJ, Cabrera-Ramírez ÁH, Ruiz-Aguirre JA, Núñez-Tapia I, Puskar M, Marques Dos Reis E, Letasiova S, Chávez-Santoscoy RA. Evaluation of Biocompatible Materials for Enhanced Mesenchymal Stem Cell Expansion: Collagen-Coated Alginate Microcarriers and PLGA Nanofibers. Biomolecules 2025; 15:345. [PMID: 40149881 PMCID: PMC11940223 DOI: 10.3390/biom15030345] [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: 01/25/2025] [Revised: 02/23/2025] [Accepted: 02/24/2025] [Indexed: 03/29/2025] Open
Abstract
Mesenchymal stem cells (MSCs) hold significant potential in regenerative medicine, tissue engineering, and cultivated meat production. However, large-scale MSC production is limited by their need for surface adherence during growth. This study evaluates two biocompatible materials-collagen-coated alginate microcarriers and polylactic-co-glycolic acid (PLGA) nanofibers-as novel growth substrates to enhance MSC proliferation. Physicochemical characterization confirmed successful collagen integration on both materials. In vitro, bone marrow-derived MSCs (bmMSCs) cultured on collagen-coated alginate microcarriers exhibited significantly enhanced growth compared to commercial microcarriers, while PLGA nanofibers supported bmMSC growth comparable to traditional growth surfaces. Scanning Electron Microscopy (SEM) revealed that bmMSCs adhered not only to the surface but also grew within the porous structure of the alginate microcarriers. Mycoplasma testing confirmed that the bmMSCs were free from contamination. Both materials were assessed for biocompatibility using ISO-10993 guidelines, demonstrating no skin or ocular irritation, supporting their potential for in situ applications in clinical and therapeutic settings. This study highlights the promise of collagen-coated alginate microcarriers and PLGA nanofibers for scalable MSC production, offering efficient, biocompatible alternatives to traditional growth surfaces in regenerative medicine and cultivated meat manufacturing. Future research should focus on optimizing these materials for larger-scale production and exploring specific applications in therapeutic and food sectors.
Collapse
Affiliation(s)
- Manuel Jaime-Rodríguez
- Tecnológico de Monterrey, School of Engineering and Science, Av. Eugenio Garza Sada 2501 Sur, Monterrey 64849, Nuevo León, Mexico
| | - María Luisa Del Prado-Audelo
- Tecnológico de Monterrey, School of Engineering and Science, Av. Eugenio Garza Sada 2501 Sur, Monterrey 64849, Nuevo León, Mexico
| | - Norma Angélica Sosa-Hernández
- Biomedical Sciences Department, Universidad Nacional Autónoma de México, Av. Universidad 3004, Coyoacán, Ciudad de Mexico 04510, Mexico
| | - Dulce Patricia Anaya-Trejo
- Tecnológico de Monterrey, School of Engineering and Science, Av. Eugenio Garza Sada 2501 Sur, Monterrey 64849, Nuevo León, Mexico
| | - Luis Jesús Villarreal-Gómez
- Engineering and Technology Science Faculty, Universidad Autónoma de Baja California, Calzada Universidad 14418, Parque Industrial, Tijuana 22424, Baja California, Mexico
| | - Ángel Humberto Cabrera-Ramírez
- Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco, A.C. Parque Científico Tecnológico de Yucatán, Km.5 Carretera, Sierra Papacal-Chuburná, Chuburná, Mérida 97302, Yucatán, Mexico
| | - Jesus Augusto Ruiz-Aguirre
- Tecnológico de Monterrey, School of Engineering and Science, Av. Eugenio Garza Sada 2501 Sur, Monterrey 64849, Nuevo León, Mexico
| | - Israel Núñez-Tapia
- Materials Research Institute, Universidad Nacional Autónoma de México, Circuito Exterior S/N, Circuito de la Investigación Científica, Coyoacán, Ciudad de Mexico 04510, Mexico
| | - Marek Puskar
- MatTek Europe, Mlynske Nivy 73, 82105 Bratislava, Slovakia
| | | | | | - Rocío Alejandra Chávez-Santoscoy
- Tecnológico de Monterrey, School of Engineering and Science, Av. Eugenio Garza Sada 2501 Sur, Monterrey 64849, Nuevo León, Mexico
| |
Collapse
|
3
|
Guo M, Qi B, Pei Z, Ni H, Ren J, Luo H, Shi H, Meng C, Yu Y, Tang Z, Xu Y, Xue Q, Li C. Therapeutic effect of low-dose BMSCs-Loaded 3D microscaffold on early osteonecrosis of the femoral head. Mater Today Bio 2025; 30:101426. [PMID: 39850243 PMCID: PMC11755031 DOI: 10.1016/j.mtbio.2024.101426] [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: 10/22/2024] [Revised: 12/14/2024] [Accepted: 12/23/2024] [Indexed: 01/25/2025] Open
Abstract
The early treatment of Osteonecrosis of Femoral Head (ONFH) remains a clinical challenge. Conventional Bone Marrow Mesenchymal Stem Cell (BMSC) injection methods often result in unsatisfactory outcomes due to mechanical cell damage, low cell survival and retention rates, inadequate cell matrix accumulation, and poor intercellular interaction. In this study, we employed a novel cell carrier material termed "3D Microscaffold" to deliver BMSCs, addressing these issues and enhancing the therapeutic effects of cell therapy for ONFH. We injected 3D microscaffold loaded with low-dose BMSCs or free high-dose BMSCs into the femoral heads of ONFH rats and assessed therapeutic effects using imaging, serology, histology, and immunohistochemistry. To understand the mechanism of efficacy, we established a co-culture model of human osteoblasts and BMSCs, followed by cell proliferation and activity detection, flow cytometry analysis, Quantitative RT-PCR, and Western blotting. Additionally, RNA sequencing was performed on femoral head tissues. Results showed that the 3D microscaffold with low-dose BMSCs had a therapeutic effect comparable to high-dose free BMSCs. Osteoblasts in the 3D microscaffold group exhibited superior phenotypes compared to the non-3D microscaffold group. Furthermore, we have, for the first time, preliminarily validated that the low-dose BMSCs-loaded 3D microscaffolds may promote the repair of femoral head necrosis through the synergistic action of the MAPK and Hippo signaling pathways.
Collapse
Affiliation(s)
- Minzheng Guo
- Chinese Academy of Medical Sciences & Peking Union Medical College, PR China
- Department of Orthopedics, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, PR China
| | - Baochuang Qi
- Department of Orthopedics, 920th Hospital of Joint Logistics Support Force, PR China
| | - Zijie Pei
- Chinese Academy of Medical Sciences & Peking Union Medical College, PR China
- Department of Orthopedics, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, PR China
| | - Haonan Ni
- Department of Orthopedics, 920th Hospital of Joint Logistics Support Force, PR China
| | - Junxiao Ren
- Department of Orthopedics, 920th Hospital of Joint Logistics Support Force, PR China
| | - Huan Luo
- Department of Orthopedics, 920th Hospital of Joint Logistics Support Force, PR China
| | - Hongxin Shi
- Department of Orthopedics, 920th Hospital of Joint Logistics Support Force, PR China
| | - Chen Meng
- Department of Orthopedics, 920th Hospital of Joint Logistics Support Force, PR China
| | - Yang Yu
- Chinese Academy of Medical Sciences & Peking Union Medical College, PR China
| | - Zhifang Tang
- Department of Orthopedics, 920th Hospital of Joint Logistics Support Force, PR China
| | - Yongqing Xu
- Department of Orthopedics, 920th Hospital of Joint Logistics Support Force, PR China
| | - Qingyun Xue
- Department of Orthopedics, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, PR China
| | - Chuan Li
- Kunming Institute of Zoology, Chinese Academy of Sciences, PR China
- Department of Orthopedics, 920th Hospital of Joint Logistics Support Force, PR China
| |
Collapse
|
4
|
Rauer SB, Stüwe L, Steinbeck L, de Toledo MAS, Fischer G, Wennemaring S, Marschick J, Koschmieder S, Wessling M, Linkhorst J. Cell Adhesion and Local Cytokine Control on Protein-Functionalized PNIPAM-co-AAc Hydrogel Microcarriers. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2404183. [PMID: 39535368 PMCID: PMC11735893 DOI: 10.1002/smll.202404183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 10/16/2024] [Indexed: 11/16/2024]
Abstract
Achieving adequate cell densities remains a major challenge in establishing economic biotechnological and biomedical processes. A possible remedy is microcarrier-based cultivation in stirred-tank bioreactors (STBR), which offers a high surface-to-volume ratio, appropriate process control, and scalability. However, despite their potential, commercial microcarriers are currently limited to material systems featuring unnatural mechanical properties and low adaptability. Because matrix stiffness and ligand presentation impact phenotypical attributes, differentiation potential, and genetic stability, biotechnological processes can significantly benefit from microcarrier systems tailorable toward cell-type specific requirements. This study introduces hydrogel particles co-polymerized from poly(N-isopropylacrylamide) (PNIPAM) and acrylic acid (AAc) as a platform technology for cell expansion. The resulting microcarriers exhibit an adjustable extracellular matrix-like softness, an adaptable gel charge, and functional carboxyl groups, allowing electrostatic and covalent coupling of cell adhesive and cell fate-modulating proteins. These features enable the attachment and growth of L929 mouse fibroblast cells in static microtiter plates and dynamic STBR cultivations while also providing vital growth factors, such as interleukin-3, to myeloblast-like 32D cells over 20 days of cultivation. The study explores the effects of different educt compositions on cell-particle interactions and reveals that PNIPAM-co-AAc microcarriers can provide both covalently coupled and diffusively released cytokine to adjacent cells.
Collapse
Affiliation(s)
- Sebastian Bernhard Rauer
- Chemical Process EngineeringRWTH Aachen UniversityForckenbeckstr. 5152074AachenGermany
- DWI ‐ Leibniz Institute for Interactive MaterialsForckenbeckstr. 5052074AachenGermany
| | - Lucas Stüwe
- Chemical Process EngineeringRWTH Aachen UniversityForckenbeckstr. 5152074AachenGermany
| | - Lea Steinbeck
- Chemical Process EngineeringRWTH Aachen UniversityForckenbeckstr. 5152074AachenGermany
| | - Marcelo Augusto Szymanski de Toledo
- Department of HematologyOncology, Hemostaseology, and Stem Cell TransplantationFaculty of MedicineRWTH Aachen University52074AachenGermany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD)52074AachenGermany
| | - Gereon Fischer
- Chemical Process EngineeringRWTH Aachen UniversityForckenbeckstr. 5152074AachenGermany
| | - Simon Wennemaring
- Chemical Process EngineeringRWTH Aachen UniversityForckenbeckstr. 5152074AachenGermany
| | - Jonas Marschick
- Chemical Process EngineeringRWTH Aachen UniversityForckenbeckstr. 5152074AachenGermany
| | - Steffen Koschmieder
- Department of HematologyOncology, Hemostaseology, and Stem Cell TransplantationFaculty of MedicineRWTH Aachen University52074AachenGermany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD)52074AachenGermany
| | - Matthias Wessling
- Chemical Process EngineeringRWTH Aachen UniversityForckenbeckstr. 5152074AachenGermany
- DWI ‐ Leibniz Institute for Interactive MaterialsForckenbeckstr. 5052074AachenGermany
| | - John Linkhorst
- Chemical Process EngineeringRWTH Aachen UniversityForckenbeckstr. 5152074AachenGermany
- Process Engineering of Electrochemical SystemsDepartment of Mechanical EngineeringTechnical University of DarmstadtOtto‐Berndt‐Str. 264287DarmstadtGermany
| |
Collapse
|
5
|
Zhang M, Huang M, Dong X, Wang Y, Zhang L, Wang Z, Cao J. Rotating cell culture system-induced injectable self-assembled microtissues with epidermal stem cells for full-thickness skin repair. PeerJ 2024; 12:e18418. [PMID: 39494298 PMCID: PMC11531757 DOI: 10.7717/peerj.18418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 10/07/2024] [Indexed: 11/05/2024] Open
Abstract
Epidermal stem cells (EpSCs) are crucial for wound healing and tissue regeneration, and traditional culture methods often lead to their inactivation. It is urgent to increase the yield of high quality EpSCs. In this study, primary EpSCs were isolated and cultured in a serum-free, feeder-free culture system. EpSCs are then expanded in a dynamic 3D environment using a rotating cell culture system (RCCS) with biodegradable porous microcarriers (MC). Over a period of 14 days, the cells self-assembled into microtissues with superior cell proliferation compared to 3D static culture. Immunofluorescence and qPCR analyses consistently showed that the stemness of the 3D microtissues was preserved, especially the COL17A1 associated with anti-aging was highly expressed in RCCS induced microtissues. In vivo experiments demonstrated that the group treated with 3D microtissues loaded with EpSCs showed enhanced early wound healing, and the injectable 3D microtissues were more conducive to maintaining cell viability and differentiation potential. Our study provides valuable insights into the dynamic 3D culture of EpSCs and introduces an injectable therapy using 3D microtissues loaded with EpSCs, which provides a new and effective approach for cell delivery and offering a promising strategy for guiding the regeneration of full-thickness skin defects.
Collapse
Affiliation(s)
- Min Zhang
- Medical School of Chinese PLA, Beijing, China
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Meng Huang
- Medical School of Chinese PLA, Beijing, China
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xixi Dong
- Department of Stomatology, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Yibo Wang
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Luyue Zhang
- Medical School of Chinese PLA, Beijing, China
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Zhaoxiang Wang
- Medical School of Chinese PLA, Beijing, China
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Junkai Cao
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
6
|
Yan L, Li D, Li S, Jiao Li J, Du G, Liu H, Zhang J, Li X, Fan Z, Jiu J, Li R, Kong N, Liu W, Du Y, Wang B. Exosomes derived from 3D-cultured MSCs alleviate knee osteoarthritis by promoting M2 macrophage polarization through miR-365a-5p and inhibiting TLR2/Myd88/NF-κB pathway. CHEMICAL ENGINEERING JOURNAL 2024; 497:154432. [DOI: 10.1016/j.cej.2024.154432] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
7
|
Zhai H, Jiang M, Zhao Y, Wang Y, Zhang H, Ji Y, Song X, Zhang J, Lv C, Li M. Three-dimensional cultured human umbilical cord mesenchymal stem cells attenuate pulmonary fibrosis by improving the balance of mitochondrial fusion and fission. Stem Cells Transl Med 2024; 13:912-926. [PMID: 39077914 PMCID: PMC11386227 DOI: 10.1093/stcltm/szae051] [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: 11/30/2023] [Accepted: 06/15/2024] [Indexed: 07/31/2024] Open
Abstract
Pulmonary fibrosis is a kind of fibrotic interstitial pneumonia with poor prognosis. Aging, environmental pollution, and coronavirus disease 2019 are considered as independent risk factors for pulmonary fibrogenesis. Consequently, the morbidity and mortality striking continues to rise in recent years. However, the clinical therapeutic efficacy is very limited and unsatisfactory. So it is necessary to develop a new effective therapeutic approach for pulmonary fibrosis. Human umbilical cord mesenchymal stem cells (hucMSCs) are considered as a promising treatment for various diseases because of their multiple differentiation and immunomodulatory function. The key bottleneck in the clinical application of hucMSCs therapy is the high-quality and large-scale production. This study used FloTrix miniSpin bioreactor, a three-dimensional (3D) cell culture system, for large-scale expansion of hucMSCs in vitro, and proved 3D cultured hucMSCs inhibited the differentiation of fibroblasts into myofibroblasts and myofibroblasts proliferation and migration, leading to slow down the development of pulmonary fibrosis. Further mechanistic studies clarified that hucMSCs reduced the amount of binding between circELP2 and miR-630, resulting in blocking YAP/TAZ translocation from cytoplasm to nucleus. This condition inhibited mitochondrial fusion and promoted mitochondrial fission, and ultimately improved fusion/fission balance and cellular homeostasis. To sum up, this work clarified the anti-fibrosis and mechanism of hucMSCs cultured from the 3D FloTrix miniSpin bioreactor. We hope to provide new ideas and new methods for the clinical transformation and industrialization of hucMSCs therapy.
Collapse
Affiliation(s)
- Huifang Zhai
- Department of Clinical Nursing, Binzhou Medical University Hospital, Binzhou Medical University, Binzhou 256603, People's Republic of China
| | - Mengqi Jiang
- Department of Cellular and Genetic Medicine, Binzhou Medical University, Yantai 264003, People's Republic of China
| | - Yaqin Zhao
- Department of Respiratory and Critical Care Medicine, Binzhou Medical University Hospital, Binzhou Medical University, Binzhou 256603, People's Republic of China
| | - Yujie Wang
- Department of Cellular and Genetic Medicine, Binzhou Medical University, Yantai 264003, People's Republic of China
| | - Haitong Zhang
- Department of Respiratory and Critical Care Medicine, Binzhou Medical University Hospital, Binzhou Medical University, Binzhou 256603, People's Republic of China
| | - Yunxia Ji
- Department of Respiratory and Critical Care Medicine, Binzhou Medical University Hospital, Binzhou Medical University, Binzhou 256603, People's Republic of China
| | - Xiaodong Song
- Department of Cellular and Genetic Medicine, Binzhou Medical University, Yantai 264003, People's Republic of China
| | - Jinjin Zhang
- Department of Cellular and Genetic Medicine, Binzhou Medical University, Yantai 264003, People's Republic of China
| | - Changjun Lv
- Department of Respiratory and Critical Care Medicine, Binzhou Medical University Hospital, Binzhou Medical University, Binzhou 256603, People's Republic of China
| | - Minge Li
- Department of Clinical Nursing, Binzhou Medical University Hospital, Binzhou Medical University, Binzhou 256603, People's Republic of China
| |
Collapse
|
8
|
Major GS, Doan VK, Longoni A, Bilek MMM, Wise SG, Rnjak-Kovacina J, Yeo GC, Lim KS. Mapping the microcarrier design pathway to modernise clinical mesenchymal stromal cell expansion. Trends Biotechnol 2024; 42:859-876. [PMID: 38320911 DOI: 10.1016/j.tibtech.2024.01.001] [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: 10/13/2023] [Revised: 01/08/2024] [Accepted: 01/08/2024] [Indexed: 02/08/2024]
Abstract
Microcarrier expansion systems show exciting potential to revolutionise mesenchymal stromal cell (MSC)-based clinical therapies by providing an opportunity for economical large-scale expansion of donor- and patient-derived cells. The poor reproducibility and efficiency of cell expansion on commercial polystyrene microcarriers have driven the development of novel microcarriers with tuneable physical, mechanical, and cell-instructive properties. These new microcarriers show innovation toward improving cell expansion outcomes, although their limited biological characterisation and compatibility with dynamic culture systems suggest the need to realign the microcarrier design pathway. Clear headway has been made toward developing infrastructure necessary for scaling up these technologies; however, key challenges remain in characterising the wholistic effects of microcarrier properties on the biological fate and function of expanded MSCs.
Collapse
Affiliation(s)
- Gretel S Major
- School of Medical Sciences, University of Sydney, Sydney, Australia
| | - Vinh K Doan
- School of Medical Sciences, University of Sydney, Sydney, Australia
| | - Alessia Longoni
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marcela M M Bilek
- School of Biomedical Engineering, University of Sydney, Sydney, Australia; School of Physics, University of Sydney, Sydney, Australia; Charles Perkins Centre, University of Sydney, Sydney, Australia; Sydney Nano Institute, University of Sydney, Sydney, Australia
| | - Steven G Wise
- School of Medical Sciences, University of Sydney, Sydney, Australia; Charles Perkins Centre, University of Sydney, Sydney, Australia
| | - Jelena Rnjak-Kovacina
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, Australia; Tyree Institute of Health Engineering, University of New South Wales, Sydney, Australia
| | - Giselle C Yeo
- Charles Perkins Centre, University of Sydney, Sydney, Australia; School of Life and Environmental Sciences, University of Sydney, Sydney, Australia.
| | - Khoon S Lim
- School of Medical Sciences, University of Sydney, Sydney, Australia; Charles Perkins Centre, University of Sydney, Sydney, Australia; Sydney Nano Institute, University of Sydney, Sydney, Australia.
| |
Collapse
|
9
|
Zhu J, Luo Q, Yang G, Xiao L. Biofabrication of Tissue-Engineered Cartilage Constructs Through Faraday Wave Bioassembly of Cell-Laden Gelatin Microcarriers. Adv Healthc Mater 2024; 13:e2304541. [PMID: 38762758 DOI: 10.1002/adhm.202304541] [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: 12/19/2023] [Revised: 04/30/2024] [Indexed: 05/20/2024]
Abstract
Acoustic biofabrication is an emerging strategy in tissue engineering due to its mild and fast manufacturing process. Herein, tissue-engineered cartilage constructs with high cell viability are fabricated from cell-laden gelatin microcarriers (GMs) through Faraday wave bioassembly, a typical acoustic "bottom-up" manufacturing process. Assembly modules are first prepared by incorporating cartilage precursor cells, the chondrogenic cell line ATDC5, or bone marrow-derived mesenchymal stem cells (BMSCs), into GMs. Patterned structures are formed by Faraday wave bioassembly of the cell-laden GMs. Due to the gentle and efficient assembly process and the protective effects of microcarriers, cells in the patterned structures maintain high activity. Subsequently, tissue-engineered cartilage constructs are obtained by inducing cell differentiation of the patterned structures. Comprehensive evaluations are conducted to verify chondrocyte differentiation and the formation of cartilage tissue constructs in terms of cell viability, morphological analysis, gene expression, and matrix production. Finally, implantation studies with a rat cartilage defect model demonstrate that these tissue-engineered cartilage constructs are beneficial for the repair of articular cartilage damage in vivo. This study provides the first biofabrication of cartilage tissue constructs using Faraday wave bioassembly, extending its application to engineering tissues with a low cell density.
Collapse
Affiliation(s)
- Jing Zhu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, China
| | - Qiuchen Luo
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, China
| | - Guang Yang
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Lin Xiao
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, China
| |
Collapse
|
10
|
Gong L, He L, Lu N, Petchakup C, Li KHH, Tay CY, Hou HW. Label-Free Single Microparticles and Cell Aggregates Sorting in Continuous Cell-Based Manufacturing. Adv Healthc Mater 2024; 13:e2304529. [PMID: 38465888 DOI: 10.1002/adhm.202304529] [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: 12/19/2023] [Revised: 03/04/2024] [Indexed: 03/12/2024]
Abstract
There is a paradigm shift in biomanufacturing toward continuous bioprocessing but cell-based manufacturing using adherent and suspension cultures, including microcarriers, hydrogel microparticles, and 3D cell aggregates, remains challenging due to the lack of efficient in-line bioprocess monitoring and cell harvesting tools. Herein, a novel label-free microfluidic platform for high throughput (≈50 particles/sec) impedance bioanalysis of biomass, cell viability, and stem cell differentiation at single particle resolution is reported. The device is integrated with a real-time piezo-actuated particle sorter based on user-defined multi-frequency impedance signatures. Biomass profiling of Cytodex-3 microcarriers seeded with adipose-derived mesenchymal stem cells (ADSCs) is first performed to sort well-seeded or confluent microcarriers for downstream culture or harvesting, respectively. Next, impedance-based isolation of microcarriers with osteogenic differentiated ADSCs is demonstrated, which is validated with a twofold increase of calcium content in sorted ADSCs. Impedance profiling of heterogenous ADSCs-encapsulated hydrogel (alginate) microparticles and 3D ADSC aggregate mixtures is also performed to sort particles with high biomass and cell viability to improve cell quality. Overall, the scalable microfluidic platform technology enables in-line sample processing from bioreactors directly and automated analysis of cell quality attributes to maximize cell yield and improve the control of cell quality in continuous cell-based manufacturing.
Collapse
Affiliation(s)
- Lingyan Gong
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore, 639798, Singapore
| | - Linwei He
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore, 639798, Singapore
| | - Nan Lu
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore, 639798, Singapore
| | - Chayakorn Petchakup
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore, 639798, Singapore
| | - King Ho Holden Li
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore, 639798, Singapore
| | - Chor Yong Tay
- School of Materials Science and Engineering, Nanyang Technological University, Singapore, 639798, Singapore
- Environmental Chemistry and Materials Centre, Nanyang Environment & Water Research Institute, Singapore, 637141, Singapore
| | - Han Wei Hou
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore, 639798, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
| |
Collapse
|
11
|
Zhu J, Luo Q, Cao T, Yang G, Xiao L. Injectable cartilage microtissues based on 3D culture using porous gelatin microcarriers for cartilage defect treatment. Regen Biomater 2024; 11:rbae064. [PMID: 38903559 PMCID: PMC11187498 DOI: 10.1093/rb/rbae064] [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: 02/19/2024] [Revised: 05/08/2024] [Accepted: 05/17/2024] [Indexed: 06/22/2024] Open
Abstract
Cartilage tissues possess an extremely limited capacity for self-repair, and current clinical surgical approaches for treating articular cartilage defects can only provide short-term relief. Despite significant advances in the field of cartilage tissue engineering, avoiding secondary damage caused by invasive surgical procedures remains a challenge. In this study, injectable cartilage microtissues were developed through 3D culture of rat bone marrow mesenchymal stem cells (BMSCs) within porous gelatin microcarriers (GMs) and induced differentiation. These microtissues were then injected for the purpose of treating cartilage defects in vivo, via a minimally invasive approach. GMs were found to be noncytotoxic and favorable for cell attachment, proliferation and migration evaluated with BMSCs. Moreover, cartilage microtissues with a considerable number of cells and abundant extracellular matrix components were obtained from BMSC-laden GMs after induction differentiation culture for 28 days. Notably, ATDC5 cells were complementally tested to verify that the GMs were conducive to cell attachment, proliferation, migration and chondrogenic differentiation. The microtissues obtained from BMSC-laden GMs were then injected into articular cartilage defect areas in rats and achieved superior performance in alleviating inflammation and repairing cartilage. These findings suggest that the use of injectable cartilage microtissues in this study may hold promise for enhancing the long-term outcomes of cartilage defect treatments while minimizing the risk of secondary damage associated with traditional surgical techniques.
Collapse
Affiliation(s)
- Jing Zhu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China
| | - Qiuchen Luo
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China
| | - Tiefeng Cao
- Department of Gynaecology, First Affiliated Hospital of Sun YatSen University, Guangzhou 510070, China
| | - Guang Yang
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Lin Xiao
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China
| |
Collapse
|
12
|
Qi Y, Wang X, Bai Z, Xu Y, Lu T, Zhu H, Zhang S, Wu Z, Liu Z, He Z, Jia W. Enhancement of the function of mesenchymal stem cells by using a GMP-grade three-dimensional hypoxic large-scale production system. Heliyon 2024; 10:e30968. [PMID: 38826705 PMCID: PMC11141262 DOI: 10.1016/j.heliyon.2024.e30968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 05/07/2024] [Accepted: 05/08/2024] [Indexed: 06/04/2024] Open
Abstract
Background Efficiently increasing the production of clinical-grade mesenchymal stem cells (MSCs) is crucial for clinical applications. Challenges with the current planar culture methods include scalability issues, labour intensity, concerns related to cell senescence, and heterogeneous responses. This study aimed to establish a large-scale production system for MSC generation. In addition, a comparative analysis of the biological differences between MSCs cultured under various conditions was conducted. Methods and materials We developed a GMP-grade three-dimensional hypoxic large-scale production (TDHLSP) system for MSCs using self-fabricated glass microcarriers and a multifunctional bioreactor. Different parameters, including cell viability, cell diameter, immunophenotype, morphology, karyotype, and tumourigenicity were assessed in MSCs cultured using different methods. Single-cell RNA sequencing (scRNA-seq) revealed pathways and genes associated with the enhanced functionality of MSCs cultured in three dimensions under hypoxic conditions (3D_Hypo MSCs). Moreover, CD142 knockdown in 3D_Hypo MSCs confirmed its in vitro functions. Results Inoculating 2 × 108 MSCs into a 2.6 L bioreactor in the TDHLSP system resulted in a final scale of 4.6 × 109 3D_Hypo MSCs by day 10. The 3D_Hypo MSCs retained characteristics of the 2D MSCs, demonstrating their genomic stability and non-tumourigenicity. Interestingly, the subpopulations of 3D_Hypo MSCs exhibited a more uniform distribution and a closer relationship than those of 2D MSCs. The heterogeneity of MSCs was strongly correlated with 'cell cycle' and 'stroma/mesenchyme', with 3D_Hypo MSCs expressing higher levels of activated stroma genes. Compared to 2D MSCs, 3D_Hypo MSCs demonstrated enhanced capabilities in blood vessel formation, TGF-β1 secretion, and inhibition of BV2 proliferation, with maintenance of Senescence-Associated β-Galactosidase (SA-β-gal) negativity. However, the enhanced functions of 3D_Hypo MSCs decreased upon the downregulation of CD142 expression. Conclusion The TDHLSP system led to a high overall production of MSCs and promoted uniform distribution of MSC clusters. This cultivation method also enhanced key cellular properties, such as angiogenesis, immunosuppression, and anti-aging. These functionally improved and uniform MSC subpopulations provide a solid basis for the clinical application of stem cell therapies.
Collapse
Affiliation(s)
- Yiyao Qi
- Institute for Regenerative Medicine, School of Life Sciences and Technology, School of Medicine, Tongji University, Shanghai, 200123, China
- National Stem Cell Translational Resource Center, Shanghai East Hospital, Tongji University, Shanghai, 200123, China
| | - Xicheng Wang
- Institute for Regenerative Medicine, School of Life Sciences and Technology, School of Medicine, Tongji University, Shanghai, 200123, China
- National Stem Cell Translational Resource Center, Shanghai East Hospital, Tongji University, Shanghai, 200123, China
| | - Zhihui Bai
- Institute for Regenerative Medicine, School of Life Sciences and Technology, School of Medicine, Tongji University, Shanghai, 200123, China
- National Stem Cell Translational Resource Center, Shanghai East Hospital, Tongji University, Shanghai, 200123, China
| | - Ying Xu
- Institute for Regenerative Medicine, School of Life Sciences and Technology, School of Medicine, Tongji University, Shanghai, 200123, China
- National Stem Cell Translational Resource Center, Shanghai East Hospital, Tongji University, Shanghai, 200123, China
| | - Tingting Lu
- Institute for Regenerative Medicine, School of Life Sciences and Technology, School of Medicine, Tongji University, Shanghai, 200123, China
- National Stem Cell Translational Resource Center, Shanghai East Hospital, Tongji University, Shanghai, 200123, China
| | - Hanyu Zhu
- Institute for Regenerative Medicine, School of Life Sciences and Technology, School of Medicine, Tongji University, Shanghai, 200123, China
- National Stem Cell Translational Resource Center, Shanghai East Hospital, Tongji University, Shanghai, 200123, China
| | - Shoumei Zhang
- Institute for Regenerative Medicine, School of Life Sciences and Technology, School of Medicine, Tongji University, Shanghai, 200123, China
- National Stem Cell Translational Resource Center, Shanghai East Hospital, Tongji University, Shanghai, 200123, China
| | - Zhihong Wu
- School of Materials Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Zhongmin Liu
- Institute for Regenerative Medicine, School of Life Sciences and Technology, School of Medicine, Tongji University, Shanghai, 200123, China
- National Stem Cell Translational Resource Center, Shanghai East Hospital, Tongji University, Shanghai, 200123, China
| | - Zhiying He
- Institute for Regenerative Medicine, School of Life Sciences and Technology, School of Medicine, Tongji University, Shanghai, 200123, China
- National Stem Cell Translational Resource Center, Shanghai East Hospital, Tongji University, Shanghai, 200123, China
| | - Wenwen Jia
- Institute for Regenerative Medicine, School of Life Sciences and Technology, School of Medicine, Tongji University, Shanghai, 200123, China
- National Stem Cell Translational Resource Center, Shanghai East Hospital, Tongji University, Shanghai, 200123, China
| |
Collapse
|
13
|
Wang X, Liang Q, Luo Y, Ye J, Yu Y, Chen F. Engineering the next generation of theranostic biomaterials with synthetic biology. Bioact Mater 2024; 32:514-529. [PMID: 38026437 PMCID: PMC10660023 DOI: 10.1016/j.bioactmat.2023.10.018] [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: 07/19/2023] [Revised: 10/06/2023] [Accepted: 10/18/2023] [Indexed: 12/01/2023] Open
Abstract
Biomaterials have evolved from inert materials to responsive entities, playing a crucial role in disease diagnosis, treatment, and modeling. However, their advancement is hindered by limitations in chemical and mechanical approaches. Synthetic biology enabling the genetically reprograming of biological systems offers a new paradigm. It has achieved remarkable progresses in cell reprogramming, engineering designer cells for diverse applications. Synthetic biology also encompasses cell-free systems and rational design of biological molecules. This review focuses on the application of synthetic biology in theranostics, which boost rapid development of advanced biomaterials. We introduce key fundamental concepts of synthetic biology and highlight frontier applications thereof, aiming to explore the intersection of synthetic biology and biomaterials. This integration holds tremendous promise for advancing biomaterial engineering with programable complex functions.
Collapse
Affiliation(s)
- Xiang Wang
- Center for Materials Synthetic Biology, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Qianyi Liang
- Center for Materials Synthetic Biology, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yixuan Luo
- Center for Materials Synthetic Biology, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Jianwen Ye
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, China
| | - Yin Yu
- Center for Materials Synthetic Biology, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Fei Chen
- Center for Materials Synthetic Biology, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| |
Collapse
|
14
|
Feng L, Li D, Tian Y, Zhao C, Sun Y, Kou X, Wu J, Wang L, Gu Q, Li W, Hao J, Hu B, Wang Y. One-step cell biomanufacturing platform: porous gelatin microcarrier beads promote human embryonic stem cell-derived midbrain dopaminergic progenitor cell differentiation in vitro and survival after transplantation in vivo. Neural Regen Res 2024; 19:458-464. [PMID: 37488911 PMCID: PMC10503631 DOI: 10.4103/1673-5374.377412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 02/07/2023] [Accepted: 04/10/2023] [Indexed: 07/26/2023] Open
Abstract
Numerous studies have shown that cell replacement therapy can replenish lost cells and rebuild neural circuitry in animal models of Parkinson's disease. Transplantation of midbrain dopaminergic progenitor cells is a promising treatment for Parkinson's disease. However, transplanted cells can be injured by mechanical damage during handling and by changes in the transplantation niche. Here, we developed a one-step biomanufacturing platform that uses small-aperture gelatin microcarriers to produce beads carrying midbrain dopaminergic progenitor cells. These beads allow midbrain dopaminergic progenitor cell differentiation and cryopreservation without digestion, effectively maintaining axonal integrity in vitro. Importantly, midbrain dopaminergic progenitor cell bead grafts showed increased survival and only mild immunoreactivity in vivo compared with suspended midbrain dopaminergic progenitor cell grafts. Overall, our findings show that these midbrain dopaminergic progenitor cell beads enhance the effectiveness of neuronal cell transplantation.
Collapse
Affiliation(s)
- Lin Feng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, China
- National Stem Cell Resource Center, Chinese Academy of Sciences, Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Da Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Yao Tian
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, China
- National Stem Cell Resource Center, Chinese Academy of Sciences, Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Chengshun Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, China
- National Stem Cell Resource Center, Chinese Academy of Sciences, Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Yun Sun
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Xiaolong Kou
- Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Jun Wu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, China
- National Stem Cell Resource Center, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Liu Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Qi Gu
- Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Wei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, China
- National Stem Cell Resource Center, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Jie Hao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, China
- National Stem Cell Resource Center, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Baoyang Hu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, China
- National Stem Cell Resource Center, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Yukai Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, China
- National Stem Cell Resource Center, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| |
Collapse
|
15
|
Kaneko M, Sato A, Ayano S, Fujita A, Kobayashi G, Ito A. Expansion of human mesenchymal stem cells on poly(vinyl alcohol) microcarriers. J Biosci Bioeng 2023; 136:407-414. [PMID: 37657971 DOI: 10.1016/j.jbiosc.2023.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 08/17/2023] [Accepted: 08/18/2023] [Indexed: 09/03/2023]
Abstract
Microcarriers provide a high surface-area-to-volume ratio that can realize high yields of cell products, including human mesenchymal stem cells (hMSCs). Here, we report a novel poly(vinyl alcohol) (PVA)-based microcarrier for hMSC expansion in suspension culture. PVA microcarriers were prepared as collagen-coated PVA hydrogels 181 μm in size and a high surface-area-to-weight ratio of 2945 cm2/g. The PVA microcarriers supported a 2.6-fold expansion of hMSCs in a 30-mL single-use stirred bioreactor after a 7 d culture period, comparable to that of commercially available microcarriers. Interestingly, we observed that hMSCs on PVA microcarriers adhered to adjacent microcarriers, resulting in the aggregation of hMSC-PVA microcarriers. Therefore, we conducted a long-term expansion culture using a bead-to-bead cell transfer method with PVA microcarriers. Fresh microcarriers were added to the cell-populated microcarriers in the bioreactor on days 7 and 14. hMSCs on PVA microcarriers continued to grow for 21 d using the bead-to-bead cell transfer method. Furthermore, magnetic PVA (PVA-mag) microcarriers were developed by loading magnetic nanoparticles into PVA microcarriers, and we demonstrated that these PVA-mag microcarriers enabled cell recovery by magnetic separation. These results suggest that these PVA microcarriers can contribute to the large-scale culture of hMSCs for regenerative medicine and cell therapy.
Collapse
Affiliation(s)
- Masahiro Kaneko
- Department of Chemical Systems Engineering, School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan
| | - Airi Sato
- College of Bioscience and Biotechnology, Chubu University, 1200 Matsumoto, Kasugai, Aichi 487-8501, Japan
| | - Satoru Ayano
- Research and Development Division, Kuraray Co., Ltd., 41 Miyukigaoka, Tsukuba, Ibaraki 305-0841, Japan
| | - Akio Fujita
- Research and Development Division, Kuraray Co., Ltd., 41 Miyukigaoka, Tsukuba, Ibaraki 305-0841, Japan
| | - Goro Kobayashi
- Research and Development Division, Kuraray Co., Ltd., 41 Miyukigaoka, Tsukuba, Ibaraki 305-0841, Japan
| | - Akira Ito
- Department of Chemical Systems Engineering, School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan.
| |
Collapse
|
16
|
Niu H, Wang BY, Wei XY, Wang YN, Zhu WH, Li WJ, Zhang Y, Wang JC. Anti-inflammatory therapeutic biomarkers identified of human bone marrow mesenchymal stem cell therapy on aging mice by serum proteomics and peptidomics study. J Proteomics 2023; 288:104979. [PMID: 37524227 DOI: 10.1016/j.jprot.2023.104979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 07/13/2023] [Accepted: 07/21/2023] [Indexed: 08/02/2023]
Abstract
Aging is accompanied by deterioration in physical condition, and creates high risks of diseases. Stem cell therapy exhibited promising potential in delaying aging. However, the unelucidated therapeutic mechanism limits future clinical application. Herein, to systematically understand the response to stem cell transfusion at the molecular level, we performed quantitative serum proteomic and peptidomics analyses in the 24-month-old aging mice model with or without mesenchymal stem cell (MSC) treatment. As a result, a total of 560 proteins and 2131 endogenous peptides were identified, among which, 6 proteins and 9 endogenous peptides derived from 6 precursor proteins were finally identified as therapeutic biomarkers after MSC transfusion on aging mice both by untargeted label-free quantification and targeted parallel reaction monitoring (PRM) quantification. Amazingly, the biological function of these differential proteins was mainly related to inflammation, which is not only the important hallmark of aging, but also the main cause of inducing aging. The reduction of these inflammatory protein content after MSC treatment further suggests the anti-inflammatory effect of MSC therapy reported elsewhere. Therefore, our study provides new evidence for the anti-inflammatory effect of MSC therapy for anti-aging and offers abundant data to support deeper investigations of the therapeutic mechanism of MSC in delaying aging.
Collapse
Affiliation(s)
- Huan Niu
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China; Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, China
| | - Bo-Yan Wang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China
| | - Xiao-Yue Wei
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China
| | - Yan-Nan Wang
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China
| | - Wen-Hui Zhu
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China
| | - Wei-Jie Li
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China
| | - Ying Zhang
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China; Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, China.
| | - Jian-Cheng Wang
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China; Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China.
| |
Collapse
|
17
|
Chen X, Li K, Chen J, Tan S. Breakthrough in large-scale production of iPSCs-derived exosomes to promote clinical applications. Front Bioeng Biotechnol 2023; 11:1257186. [PMID: 37691905 PMCID: PMC10484304 DOI: 10.3389/fbioe.2023.1257186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 08/15/2023] [Indexed: 09/12/2023] Open
Affiliation(s)
| | | | | | - Songwen Tan
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, China
| |
Collapse
|
18
|
Wang Z, Zhang X, Xue L, Wang G, Li X, Chen J, Xu R, Xu T. A controllable gelatin-based microcarriers fabrication system for the whole procedures of MSCs amplification and tissue engineering. Regen Biomater 2023; 10:rbad068. [PMID: 37638061 PMCID: PMC10458456 DOI: 10.1093/rb/rbad068] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 07/06/2023] [Accepted: 07/30/2023] [Indexed: 08/29/2023] Open
Abstract
Biopolymer microbeads present substantial benefits for cell expansion, tissue engineering, and drug release applications. However, a fabrication system capable of producing homogeneous microspheres with high precision and controllability for cell proliferation, passaging, harvesting and downstream application is limited. Therefore, we developed a co-flow microfluidics-based system for the generation of uniform and size-controllable gelatin-based microcarriers (GMs) for mesenchymal stromal cells (MSCs) expansion and tissue engineering. Our evaluation of GMs revealed superior homogeneity and efficiency of cellular attachment, expansion and harvest, and MSCs expanded on GMs exhibited high viability while retaining differentiation multipotency. Optimization of passaging and harvesting protocols was achieved through the addition of blank GMs and treatment with collagenase, respectively. Furthermore, we demonstrated that MSC-loaded GMs were printable and could serve as building blocks for tissue regeneration scaffolds. These results suggested that our platform held promise for the fabrication of uniform GMs with downstream application of MSC culture, expansion and tissue engineering.
Collapse
Affiliation(s)
- Zixian Wang
- Precision Medicine and Healthcare Research Center, Tsinghua-Berkeley Shenzhen Institute (TBSI), Tsinghua University, Shenzhen 518055, People’s Republic of China
| | - Xiuxiu Zhang
- Precision Medicine and Healthcare Research Center, Tsinghua-Berkeley Shenzhen Institute (TBSI), Tsinghua University, Shenzhen 518055, People’s Republic of China
| | - Limin Xue
- Department of Research and Development, Huaqing Zhimei (Shenzhen) Biotechnology Co., Ltd., Shenzhen 518107, People’s Republic of China
| | - Gangwei Wang
- Department of Emergency, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, People’s Republic of China
| | - Xinda Li
- Department of Neurosurgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu 610072, People’s Republic of China
| | - Jianwei Chen
- Bio-intelligent Manufacturing and Living Matter Bioprinting Center, Research Institute of Tsinghua University in Shenzhen, Tsinghua University, Shenzhen 518057, People’s Republic of China
| | - Ruxiang Xu
- Department of Neurosurgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu 610072, People’s Republic of China
| | - Tao Xu
- Precision Medicine and Healthcare Research Center, Tsinghua-Berkeley Shenzhen Institute (TBSI), Tsinghua University, Shenzhen 518055, People’s Republic of China
- Department of Neurosurgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu 610072, People’s Republic of China
- Bio-intelligent Manufacturing and Living Matter Bioprinting Center, Research Institute of Tsinghua University in Shenzhen, Tsinghua University, Shenzhen 518057, People’s Republic of China
| |
Collapse
|
19
|
Gao T, Zhao X, Hao J, Tian Y, Ma H, Liu W, An B, Sun F, Liu S, Guo B, Niu S, Li Z, Wang C, Wang Y, Feng G, Wang L, Li W, Wu J, Guo M, Zhou Q, Gu Q. A scalable culture system incorporating microcarrier for specialised mesenchymal stem cells from human embryonic stem cells. Mater Today Bio 2023; 20:100662. [PMID: 37214547 PMCID: PMC10196860 DOI: 10.1016/j.mtbio.2023.100662] [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: 03/07/2023] [Revised: 04/20/2023] [Accepted: 05/05/2023] [Indexed: 05/24/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) derived from human embryonic stem cells (hESCs) are a desirable cell source for cell therapy owing to their capacity to be produced stably and homogeneously in large quantities. However, a scalable culture system for hPSC-derived MSCs is urgently needed to meet the cell quantity and quality requirements of practical clinical applications. In this study, we developed a new microcarrier with hyaluronic acid (HA) as the core material, which allowed scalable serum-free suspension culture of hESC-derived MSCs (IMRCs). We used optimal microcarriers with a coating collagen concentration of 100 μg/mL or concave-structured surface (cHAMCs) for IMRC amplification in a stirred bioreactor, expanding IMRCs within six days with the highest yield of over one million cells per milliliter. In addition, the harvested cells exhibited high viability, immunomodulatory and regenerative therapeutic promise comparable to monolayer cultured MSCs while showing more increased secretion of extracellular matrix (ECM), particularly collagen-related proteins. In summary, we have established a scalable culture system for hESC-MSCs, providing novel approaches for future cell therapies.
Collapse
Affiliation(s)
- Tingting Gao
- State Key Laboratory of Stem Cell and Reproductive Biology, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiyuan Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jie Hao
- National Stem Cell Resource Center, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yao Tian
- State Key Laboratory of Stem Cell and Reproductive Biology, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Huike Ma
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Wenjing Liu
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Bin An
- State Key Laboratory of Stem Cell and Reproductive Biology, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Faguo Sun
- State Key Laboratory of Stem Cell and Reproductive Biology, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shasha Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Baojie Guo
- State Key Laboratory of Stem Cell and Reproductive Biology, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuaishuai Niu
- National Stem Cell Resource Center, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Zhongwen Li
- State Key Laboratory of Stem Cell and Reproductive Biology, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Chenxin Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Yukai Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guihai Feng
- State Key Laboratory of Stem Cell and Reproductive Biology, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Liu Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jun Wu
- State Key Laboratory of Stem Cell and Reproductive Biology, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Meijin Guo
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Qi Zhou
- State Key Laboratory of Stem Cell and Reproductive Biology, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qi Gu
- State Key Laboratory of Stem Cell and Reproductive Biology, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
20
|
Li J, Wu Y, Yao X, Tian Y, Sun X, Liu Z, Ye X, Wu C. Preclinical Research of Stem Cells: Challenges and Progress. Stem Cell Rev Rep 2023:10.1007/s12015-023-10528-y. [PMID: 37097496 DOI: 10.1007/s12015-023-10528-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/06/2023] [Indexed: 04/26/2023]
Abstract
In recent years, great breakthroughs have been made in basic research and clinical applications of stem cells in regenerative medicine and other fields, which continue to inspire people to explore the field of stem cells. With nearly unlimited self-renewal ability, stem cells can generate at least one type of highly differentiated daughter cell, which provides broad development prospects for the treatment of human organ damage and other diseases. In the field of stem cell research, related technologies for inducing or isolating stem cells are relatively mature, and a variety of stable stem cell lines have been successfully constructed. To realize the full clinical application of stem cells as soon as possible, it is more and more important to further optimize each stage of stem cell research while conforming to Current Good Manufacture Practices (cGMP) standards. Here, we synthesized recent developments in stem cell research and focus on the introduction of xenogenicity in the preclinical research process and the remaining problems of various cell bioreactors. Our goal is to promote the development of technologies for xeno-free culture and clinical expansion of stem cells through in-depth discussion of current research. This review will provide new insight into stem cell research protocols and will contribute to the creation of efficient and stable stem cell expansion systems.
Collapse
Affiliation(s)
- Jinhu Li
- School of Pharmacy, School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yurou Wu
- School of Pharmacy, School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiang Yao
- School of Pharmacy, School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yao Tian
- School of Pharmacy, School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xue Sun
- School of Pharmacy, School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zibo Liu
- School of Pharmacy, School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xun Ye
- School of Pharmacy, School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chunjie Wu
- Innovative Institute of Chinese Medicine and Pharmacy/Academy for Interdiscipline, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
21
|
Li Y, He M, Zhang W, Liu W, Xu H, Yang M, Zhang H, Liang H, Li W, Wu Z, Fu W, Xu S, Liu X, Fan S, Zhou L, Wang C, Zhang L, Li Y, Gu J, Yin J, Zhang Y, Xia Y, Mao X, Cheng T, Shi J, Du Y, Gao Y. Expansion of human megakaryocyte-biased hematopoietic stem cells by biomimetic Microniche. Nat Commun 2023; 14:2207. [PMID: 37072407 PMCID: PMC10113370 DOI: 10.1038/s41467-023-37954-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 04/04/2023] [Indexed: 04/20/2023] Open
Abstract
Limited numbers of available hematopoietic stem cells (HSCs) limit the widespread use of HSC-based therapies. Expansion systems for functional heterogenous HSCs remain to be optimized. Here, we present a convenient strategy for human HSC expansion based on a biomimetic Microniche. After demonstrating the expansion of HSC from different sources, we find that our Microniche-based system expands the therapeutically attractive megakaryocyte-biased HSC. We demonstrate scalable HSC expansion by applying this strategy in a stirred bioreactor. Moreover, we identify that the functional human megakaryocyte-biased HSCs are enriched in the CD34+CD38-CD45RA-CD90+CD49f lowCD62L-CD133+ subpopulation. Specifically, the expansion of megakaryocyte-biased HSCs is supported by a biomimetic niche-like microenvironment, which generates a suitable cytokine milieu and supplies the appropriate physical scaffolding. Thus, beyond clarifying the existence and immuno-phenotype of human megakaryocyte-biased HSC, our study demonstrates a flexible human HSC expansion strategy that could help realize the strong clinical promise of HSC-based therapies.
Collapse
Affiliation(s)
- Yinghui Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Mei He
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Wenshan Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Wei Liu
- Department of Biomedical Engineering, School of Medicine, Tsinghua-PKU Center for Life Sciences, Tsinghua University, 100084, Beijing, China
- Beijing CytoNiche Biotechnology Co. Ltd., 100195, Beijing, China
| | - Hui Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Ming Yang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Hexiao Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Haiwei Liang
- Department of Biomedical Engineering, School of Medicine, Tsinghua-PKU Center for Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Wenjing Li
- Department of Biomedical Engineering, School of Medicine, Tsinghua-PKU Center for Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Zhaozhao Wu
- Department of Biomedical Engineering, School of Medicine, Tsinghua-PKU Center for Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Weichao Fu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Shiqi Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Xiaolei Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Sibin Fan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Liwei Zhou
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Chaoqun Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Lele Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Yafang Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Jiali Gu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Jingjing Yin
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Yiran Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Yonghui Xia
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Xuemei Mao
- Nankai Hospital, Tianjin Hospital of Integrated Traditional Chinese and Western Medicine, Tianjin, 300100, China
| | - Tao Cheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China.
- Tianjin Institutes of Health Science, Tianjin, 301600, China.
| | - Jun Shi
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China.
- Tianjin Institutes of Health Science, Tianjin, 301600, China.
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Tsinghua-PKU Center for Life Sciences, Tsinghua University, 100084, Beijing, China.
- Beijing CytoNiche Biotechnology Co. Ltd., 100195, Beijing, China.
| | - Yingdai Gao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, PUMC Department of Stem Cell and Regenerative Medicine, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China.
- Tianjin Institutes of Health Science, Tianjin, 301600, China.
| |
Collapse
|
22
|
Handral HK, Wyrobnik TA, Lam ATL. Emerging Trends in Biodegradable Microcarriers for Therapeutic Applications. Polymers (Basel) 2023; 15:polym15061487. [PMID: 36987266 PMCID: PMC10057597 DOI: 10.3390/polym15061487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/13/2023] [Accepted: 03/14/2023] [Indexed: 03/19/2023] Open
Abstract
Microcarriers (MCs) are adaptable therapeutic instruments that may be adjusted to specific therapeutic uses, making them an appealing alternative for regenerative medicine and drug delivery. MCs can be employed to expand therapeutic cells. MCs can be used as scaffolds for tissue engineering, as well as providing a 3D milieu that replicates the original extracellular matrix, facilitating cell proliferation and differentiation. Drugs, peptides, and other therapeutic compounds can be carried by MCs. The surface of the MCs can be altered, to improve medication loading and release, and to target specific tissues or cells. Allogeneic cell therapies in clinical trials require enormous volumes of stem cells, to assure adequate coverage for several recruitment locations, eliminate batch to batch variability, and reduce production costs. Commercially available microcarriers necessitate additional harvesting steps to extract cells and dissociation reagents, which reduces cell yield and quality. To circumvent such production challenges, biodegradable microcarriers have been developed. In this review, we have compiled key information relating to biodegradable MC platforms, for generating clinical-grade cells, that permit cell delivery at the target site without compromising quality or cell yields. Biodegradable MCs could also be employed as injectable scaffolds for defect filling, supplying biochemical signals for tissue repair and regeneration. Bioinks, coupled with biodegradable microcarriers with controlled rheological properties, might improve bioactive profiles, while also providing mechanical stability to 3D bioprinted tissue structures. Biodegradable materials used for microcarriers have the ability to solve in vitro disease modeling, and are advantageous to the biopharmaceutical drug industries, because they widen the spectrum of controllable biodegradation and may be employed in a variety of applications.
Collapse
Affiliation(s)
- Harish K. Handral
- Stem Cell Bioprocessing, Bioprocessing Technology Institute, A*STAR, Singapore 138668, Singapore
- Correspondence:
| | - Tom Adam Wyrobnik
- Stem Cell Bioprocessing, Bioprocessing Technology Institute, A*STAR, Singapore 138668, Singapore
- Department of Biochemical Engineering, University College London, Gower Street, London WC1E 6BT, UK
| | - Alan Tin-Lun Lam
- Stem Cell Bioprocessing, Bioprocessing Technology Institute, A*STAR, Singapore 138668, Singapore
| |
Collapse
|
23
|
Xiang Y, Yan J, Bao X, Gleadall A, Roach P, Sun T. Evaluation of Polymeric Particles for Modular Tissue Cultures in Developmental Engineering. Int J Mol Sci 2023; 24:ijms24065234. [PMID: 36982306 PMCID: PMC10049291 DOI: 10.3390/ijms24065234] [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: 02/14/2023] [Revised: 03/03/2023] [Accepted: 03/07/2023] [Indexed: 03/30/2023] Open
Abstract
Developmental engineering (DE) aims to culture mammalian cells on corresponding modular scaffolds (scale: micron to millimeter), then assemble these into functional tissues imitating natural developmental biology processes. This research intended to investigate the influences of polymeric particles on modular tissue cultures. When poly(methyl methacrylate) (PMMA), poly(lactic acid) (PLA) and polystyrene (PS) particles (diameter: 5-100 µm) were fabricated and submerged in culture medium in tissue culture plastics (TCPs) for modular tissue cultures, the majority of adjacent PMMA, some PLA but no PS particles aggregated. Human dermal fibroblasts (HDFs) could be directly seeded onto large (diameter: 30-100 µm) PMMA particles, but not small (diameter: 5-20 µm) PMMA, nor all the PLA and PS particles. During tissue cultures, HDFs migrated from the TCPs surfaces onto all the particles, while the clustered PMMA or PLA particles were colonized by HDFs into modular tissues with varying sizes. Further comparisons revealed that HDFs utilized the same cell bridging and stacking strategies to colonize single or clustered polymeric particles, and the finely controlled open pores, corners and gaps on 3D-printed PLA discs. These observed cell-scaffold interactions, which were then used to evaluate the adaptation of microcarrier-based cell expansion technologies for modular tissue manufacturing in DE.
Collapse
Affiliation(s)
- Yu Xiang
- Department of Materials, Loughborough University, Epinal Way, Loughborough LE11 3TU, UK
| | - Jiongyi Yan
- Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Epinal Way, Loughborough LE11 3TU, UK
| | - Xujin Bao
- Department of Materials, Loughborough University, Epinal Way, Loughborough LE11 3TU, UK
| | - Andrew Gleadall
- Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Epinal Way, Loughborough LE11 3TU, UK
| | - Paul Roach
- Department of Chemistry, Loughborough University, Epinal Way, Loughborough LE11 3TU, UK
| | - Tao Sun
- Department of Chemical Engineering, Loughborough University, Epinal Way, Loughborough LE11 3TU, UK
| |
Collapse
|
24
|
Koh B, Sulaiman N, Fauzi MB, Law JX, Ng MH, Yuan TL, Azurah AGN, Mohd Yunus MH, Idrus RBH, Yazid MD. A Three-Dimensional Xeno-Free Culture Condition for Wharton's Jelly-Mesenchymal Stem Cells: The Pros and Cons. Int J Mol Sci 2023; 24:ijms24043745. [PMID: 36835154 PMCID: PMC9960744 DOI: 10.3390/ijms24043745] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 01/19/2023] [Accepted: 01/22/2023] [Indexed: 02/15/2023] Open
Abstract
Xeno-free three-dimensional cultures are gaining attention for mesenchymal stem cell (MSCs) expansion in clinical applications. We investigated the potential of xeno-free serum alternatives, human serum and human platelet lysate, to replace the current conventional use of foetal bovine serum for subsequent MSCs microcarrier cultures. In this study, Wharton's Jelly MSCs were cultured in nine different media combinations to identify the best xeno-free culture media for MSCs culture. Cell proliferation and viability were identified, and the cultured MSCs were characterised in accordance with the minimal criteria for defining multipotent mesenchymal stromal cells by the International Society for Cellular Therapy (ISCT). The selected culture media was then used in the microcarrier culture of MSCs to determine the potential of a three-dimensional culture system in the expansion of MSCs for future clinical applications, and to identify the immunomodulatory potential of cultured MSCs. Low Glucose DMEM (LG) + Human Platelet (HPL) lysate media appeared to be good candidates for replacing conventional MSCs culture media in our monolayer culture system. MSCs cultured in LG-HPL achieved high cell yield, with characteristics that remained as described by ISCT, although the overall mitochondrial activity of the cells was lower than the control and the subsequent effects remained unknown. MSC microcarrier culture, on the other hand, showed comparable cell characteristics with monolayer culture, yet had stagnated cell proliferation, which is potentially due to the inactivation of FAK. Nonetheless, both the MSCs monolayer culture and the microcarrier culture showed high suppressive activity on TNF-α, and only the MSC microcarrier culture has a better suppression of IL-1 secretion. In conclusion, LG-HPL was identified as a good xeno-free media for WJMSCs culture, and although further mechanistic research is needed, the results show that the xeno-free three-dimensional culture maintained MSC characteristics and improved immunomodulatory activities, suggesting the potential of translating the monolayer culture into this culture system in MSC expansion for future clinical application.
Collapse
Affiliation(s)
- Benson Koh
- Centre for Tissue Engineering & Regenerative Medicine, Faculty of Medicine, Jalan Yaacob Latif, Cheras, Kuala Lumpur 56000, Malaysia
- Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras, Kuala Lumpur 56000, Malaysia
- Ming Medical Sdn Bhd, D3-3 (2nd Floor), Block D3 Dana 1 Commercial Centre, Jalan PJU 1a/46, Petaling Jaya 47301, Malaysia
| | - Nadiah Sulaiman
- Centre for Tissue Engineering & Regenerative Medicine, Faculty of Medicine, Jalan Yaacob Latif, Cheras, Kuala Lumpur 56000, Malaysia
- Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras, Kuala Lumpur 56000, Malaysia
| | - Mh Busra Fauzi
- Centre for Tissue Engineering & Regenerative Medicine, Faculty of Medicine, Jalan Yaacob Latif, Cheras, Kuala Lumpur 56000, Malaysia
- Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras, Kuala Lumpur 56000, Malaysia
| | - Jia Xian Law
- Centre for Tissue Engineering & Regenerative Medicine, Faculty of Medicine, Jalan Yaacob Latif, Cheras, Kuala Lumpur 56000, Malaysia
- Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras, Kuala Lumpur 56000, Malaysia
| | - Min Hwei Ng
- Centre for Tissue Engineering & Regenerative Medicine, Faculty of Medicine, Jalan Yaacob Latif, Cheras, Kuala Lumpur 56000, Malaysia
- Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras, Kuala Lumpur 56000, Malaysia
| | - Too Lih Yuan
- Centre for Tissue Engineering & Regenerative Medicine, Faculty of Medicine, Jalan Yaacob Latif, Cheras, Kuala Lumpur 56000, Malaysia
- Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras, Kuala Lumpur 56000, Malaysia
| | - Abdul Ghani Nur Azurah
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras, Kuala Lumpur 56000, Malaysia
| | - Mohd Heikal Mohd Yunus
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras, Kuala Lumpur 56000, Malaysia
| | - Ruszymah Bt Hj Idrus
- Centre for Tissue Engineering & Regenerative Medicine, Faculty of Medicine, Jalan Yaacob Latif, Cheras, Kuala Lumpur 56000, Malaysia
- Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras, Kuala Lumpur 56000, Malaysia
| | - Muhammad Dain Yazid
- Centre for Tissue Engineering & Regenerative Medicine, Faculty of Medicine, Jalan Yaacob Latif, Cheras, Kuala Lumpur 56000, Malaysia
- Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras, Kuala Lumpur 56000, Malaysia
- Correspondence: ; Tel.: +60-3-9145-6995
| |
Collapse
|
25
|
Wei X, Li R, Li X, Wang B, Huang J, Mu H, Zhang Q, Zhang Z, Ru Y, Wu X, Qiu Y, Ye Y, Feng Y, Wang S, Chen H, Yi C, Wang J. iPSCs-derived mesenchymal stromal cells mitigate anxiety and neuroinflammation in aging female mice. Int J Biochem Cell Biol 2023; 155:106347. [PMID: 36565990 DOI: 10.1016/j.biocel.2022.106347] [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: 10/04/2022] [Revised: 11/22/2022] [Accepted: 12/18/2022] [Indexed: 12/24/2022]
Abstract
Perimenopause is a natural transition to menopause, when hormone disturbance can result in both short-term mental disorders, such as anxiety, and long-term neuroinflammation due to blood-brain barrier (BBB) impairment, which may lead to more serious neurological disorders later on, such as dementia. Effective treatments may prevent both short-term and long-term neurological sequela, which formed the aim of this study. In aged female C57BL/6 mice (16-18 months of age), mesenchymal stromal cells (MSCs) differentiated from human-induced pluripotent stem cells (iPSCs), were administered via tail vein injection. Mice showed increased blood estrogen levels, alleviated anxiety and neuroinflammation, and improved BBB integrity. Interestingly, transplanted MSCs were located close to ovarian sympathetic nerves and decreased ovarian norepinephrine levels, which in turn increased ovarian estrogen secretion. Moreover, the administration of anastrozole, an inhibitor of estrogen synthesis, diminished the therapeutic effects of MSCs in vivo, suggesting the effect to be estrogen-dependent. In vitro study confirmed the impact of MSCs on sympathetic nerves via mitochondria exchange. In conclusion, iPSC-derived MSCs may provide a novel option to manage perimenopause-related hormonal dysregulation and neurological disorders during the female aging process.
Collapse
Affiliation(s)
- Xiaoyue Wei
- Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China; Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510060, China
| | - Ruijie Li
- Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China
| | - Xiangyu Li
- Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China
| | - Boyan Wang
- Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China
| | - Jianyang Huang
- Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China
| | - Hanyiqi Mu
- Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China
| | - Qinmu Zhang
- Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China
| | - Ziyuan Zhang
- Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China
| | - Yifei Ru
- Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China
| | - Xinxiang Wu
- Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China
| | - Yuan Qiu
- Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China
| | - Yanchen Ye
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Yuanyuan Feng
- Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China
| | - Shiyu Wang
- Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China
| | - Hui Chen
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Chenju Yi
- Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China; Guangdong Provincal Key Laboratory of Brain Function and Disease, Guangzhou 510080, China.
| | - Jiancheng Wang
- Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China.
| |
Collapse
|
26
|
Zhao Z, Wang Y, Yin B, Li X, Hao R, Li Z, Li P, Han M, Ding L, Li Z, Zhu H. Defect-adaptive Stem-cell-microcarrier Construct Promotes Tissue Repair in Rabbits with Knee Cartilage Defects. Stem Cell Rev Rep 2023; 19:201-212. [PMID: 35900693 DOI: 10.1007/s12015-022-10421-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/23/2022] [Indexed: 01/29/2023]
Abstract
Although various reconstruction techniques are available for cartilage defects, the repair effects and conveniences remain to be further improved due to the limited regenerative capacity of cartilaginous tissues and difficulties in seamlessly fulfilling irregularly shaped defects. In the current study, we explored the repair efficacy of stem cell microcarrier construct (microcarriers loaded with human chondrogenic progenitor cells or bone marrow mesenchymal stem cells) in cartilage defect models. A total of 39 healthy New Zealand white rabbits were included, and femoral trochlear cartilage defect models were established (n = 33). Stem cell microcarrier constructs were implanted into cartilage defects (n = 6), the maintenance conditions of the implanted constructs were observed on days 4, 8, and 30 post implantation (n = 3). Gross observation and pathological analysis were performed to assay the reconstitution of cartilage defects at 12 weeks post-cartilage defect repair(n = 6). The microcarriers could fill the defect model with good plasticity to integrate well with the boundary native normal cartilage. At 3 months after implantation, the defects were filled with fibrous cartilage tissues in the microcarrier without stem cells group. In the microcarrier loaded with BMSCs group, newly formed tissue with a similar appearance of boundary cartilage fulfilled the defects, but the surface was not completely smooth. Promisingly, the defects were almost completely filled with newly regenerated cartilaginous tissues, which had a smooth appearance similar to that of normal cartilage in the microcarrier loaded with CPCs group. These results suggest the feasibility of stem cell microcarrier construct in repairing cartilage defects, indicating promising clinical application prospects.
Collapse
Affiliation(s)
- Zhidong Zhao
- Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China.,Chinese People's Liberation Army (PLA) General Hospital, Chinese PLA Medical School, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Yuxing Wang
- Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China.,Chinese People's Liberation Army (PLA) General Hospital, Chinese PLA Medical School, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Bofeng Yin
- Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Xiaotong Li
- Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Ruicong Hao
- Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China.,Graduate School of Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Zhiling Li
- Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Peilin Li
- Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Mengyue Han
- Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China.,Graduate School of Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Li Ding
- Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China. .,Air Force Medical Center, PLA, No.30 Fucheng Road, Beijing, 100142, China.
| | - Zhongli Li
- Chinese People's Liberation Army (PLA) General Hospital, Chinese PLA Medical School, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China.
| | - Heng Zhu
- Beijing Institute of Radiation Medicine, No. 27 Taiping Road, Haidian District, Beijing, 100850, China. .,Graduate School of Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui, China.
| |
Collapse
|
27
|
Ding SL, Liu X, Zhao XY, Wang KT, Xiong W, Gao ZL, Sun CY, Jia MX, Li C, Gu Q, Zhang MZ. Microcarriers in application for cartilage tissue engineering: Recent progress and challenges. Bioact Mater 2022; 17:81-108. [PMID: 35386447 PMCID: PMC8958326 DOI: 10.1016/j.bioactmat.2022.01.033] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 12/11/2022] Open
Abstract
Successful regeneration of cartilage tissue at a clinical scale has been a tremendous challenge in the past decades. Microcarriers (MCs), usually used for cell and drug delivery, have been studied broadly across a wide range of medical fields, especially the cartilage tissue engineering (TE). Notably, microcarrier systems provide an attractive method for regulating cell phenotype and microtissue maturations, they also serve as powerful injectable carriers and are combined with new technologies for cartilage regeneration. In this review, we introduced the typical methods to fabricate various types of microcarriers and discussed the appropriate materials for microcarriers. Furthermore, we highlighted recent progress of applications and general design principle for microcarriers. Finally, we summarized the current challenges and promising prospects of microcarrier-based systems for medical applications. Overall, this review provides comprehensive and systematic guidelines for the rational design and applications of microcarriers in cartilage TE.
Collapse
Affiliation(s)
- Sheng-Long Ding
- Center of Foot and Ankle Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Xin Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xi-Yuan Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Ke-Tao Wang
- Center of Foot and Ankle Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Wei Xiong
- Center of Foot and Ankle Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Zi-Li Gao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Cheng-Yi Sun
- Center of Foot and Ankle Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Min-Xuan Jia
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Cheng Li
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, 100083, China
| | - Qi Gu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regeneration, University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Ming-Zhu Zhang
- Center of Foot and Ankle Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| |
Collapse
|
28
|
Wu R, Li H, Sun C, Liu J, Chen D, Yu H, Huang Z, Lin S, Chen Y, Zheng Q. Exosome-based strategy for degenerative disease in orthopedics: Recent progress and perspectives. J Orthop Translat 2022; 36:8-17. [PMID: 35891923 PMCID: PMC9283806 DOI: 10.1016/j.jot.2022.05.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 05/07/2022] [Accepted: 05/19/2022] [Indexed: 02/09/2023] Open
Abstract
BACKGROUND Degenerative diseases in orthopaedics have become a significant global public health issue with the aging of the population worldwide. The traditional medical interventions, including physical therapy, pharmacological therapy and even surgery, hardly work to modify degenerative progression. Stem cell-based therapy is widely accepted to treat degenerative orthopaedic disease effectively but possesses several limitations, such as the need for strict monitoring of production and storage and the potential risks of tumorigenicity and immune rejection in clinical translation. Furthermore, the ethical issues surrounding the acquisition of embryonic stem cells are also broadly concerned. Exosome-based therapy has rapidly grown in popularity in recent years and is regarded as an ideal alternative to stem cell-based therapy, offering a promise to achieve 'cell-free' tissue regeneration. METHODS Traditionally, the native exosomes extracted from stem cells are directly injected into the injured site to promote tissue regeneration. Recently, several modified exosome-based strategies were developed to overcome the limitations of native exosomes, which include mainly exogenous molecule loading and exosome delivery through scaffolds. In this paper, a systematic review of the exosome-based strategy for degenerative disease in orthopaedics is presented. RESULTS Treatment strategies based on the native exosomes are effective but with several disadvantages such as rapid diffusion and insufficient and fluctuating functional contents. The modified exosome-based strategies can better match the requirements of the regeneration in some complex healing processes. CONCLUSION Exosome-based strategies hold promise to manage degenerative disease in orthopaedics prior to patients reaching the advanced stage of disease in the future. The timely summary and highlights offered herein could provide a research perspective to promote the development of exosome-based therapy, facilitating the clinical translation of exosomes in orthopaedics. TRANSLATIONAL POTENTIAL OF THIS ARTICLE Exosome-based therapy is superior in anti-senescence and anti-inflammatory effects and possesses lower risks of tumorigenicity and immune rejection relative to stem cell-based therapy. Exosome-based therapy is regarded as an ideal alternative to stem cell-based therapy, offering a promise to achieve 'cell-free' tissue regeneration.
Collapse
Affiliation(s)
- Rongjie Wu
- Department of Orthopedics, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, PR China
- Shantou University Medical College, Shantou, China
| | - Haotao Li
- Department of Orthopedics, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, PR China
- Shantou University Medical College, Shantou, China
| | - Chuanwei Sun
- Department of Burn and Wound Repair Surgery and Research Department of Medical Science, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, PR China
| | - Jialin Liu
- Rehabilitation Center, Shengjing Hospital Affiliated to China Medical University, Shenyang, Liaoning, PR China
| | - Duanyong Chen
- Department of Orthopedics, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, PR China
| | - Haiyang Yu
- Department of Orthopedics, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, PR China
| | - Zena Huang
- Department of General Medicine, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Sien Lin
- Department of Orthopaedics & Traumatology, Stem Cells and Regenerative Medicine Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong Special Administrative Region of China
| | - Yuanfeng Chen
- Department of Orthopedics, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, PR China
- Research Department of Medical Science, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, PR China
| | - Qiujian Zheng
- Department of Orthopedics, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, PR China
- Southern Medical University, Guangzhou, PR China
| |
Collapse
|
29
|
Mawji I, Roberts EL, Dang T, Abraham B, Kallos MS. Challenges and Opportunities in Downstream Separation Processes for Mesenchymal Stromal Cells Cultured in Microcarrier-based Stirred Suspension Bioreactors. Biotechnol Bioeng 2022; 119:3062-3078. [PMID: 35962467 DOI: 10.1002/bit.28210] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 07/27/2022] [Accepted: 08/11/2022] [Indexed: 11/08/2022]
Abstract
Mesenchymal stromal cells (MSC) are a promising platform for regenerative medicine applications because of their multi-lineage differentiation abilities and ease of collection, isolation, and growth ex-vivo. To meet the demand for clinical applications, large scale manufacturing will be required using three-dimension culture platforms in vessels such as stirred suspension bioreactors. As MSCs are an adherent cell type, microcarriers are added to the culture to increase the available surface area for attachment and growth. Although extensive research has been performed on efficiently culturing MSCs using microcarriers, challenges persist in downstream processing including harvesting, filtration, and volume reduction which all play a critical role for the translation of cell therapies to the clinic. The objective of this review is to assess the current state of downstream technologies available for microcarrier-based MSC cultures. This includes a review of current research within the three stages: harvesting, filtration, and volume reduction. Using this information, a downstream process for MSCs is proposed which can be applied for a wide range of applications. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Inaara Mawji
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada.,Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada
| | - Erin L Roberts
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada.,Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada
| | - Tiffany Dang
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada.,Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada
| | - Brett Abraham
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada.,Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada
| | - Michael S Kallos
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada.,Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
30
|
Zhang Y, Na T, Zhang K, Yang Y, Xu H, Wei L, Xu L, Yan X, Liu W, Liu G, Wang B, Meng S, Du Y. GMP-grade microcarrier and automated closed industrial scale cell production platform for culture of MSCs. J Tissue Eng Regen Med 2022; 16:934-944. [PMID: 35929499 DOI: 10.1002/term.3341] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/23/2022] [Accepted: 07/11/2022] [Indexed: 11/08/2022]
Abstract
Efficient and large-scale expansion of mesenchymal stem/stromal cells (MSCs) has always been a formidable challenge to researchers in cell-based therapies and regenerative medicine. To reconcile major drawbacks of 2D planar culturing system, we innovatively developed an automated closed industrial scale cell production (ACISCP) platform based on GMP-grade microcarrier for culture of umbilical cord-mesenchymal stem/stromal cells (UCMSCs), in accordance with the criteria of stem cell bank. ACISCP system is a fully closed system, which employs different models of vivaSPIN bioreactors (CytoNiche Biotech, China) for scale-up cell culture and vivaPREP (CytoNiche Biotech, China) for automated cell harvesting and cell dosage preparation. To realize industrial scale expansion of UCMSCs, a three-stage expansion was conducted with 1 L, 5 and 15 L vivaSPIN bioreactors. Using 3D TableTrix® and ACISCP system, we inoculated 1.5 × 107 of UCMSCs into 1 L vivaSPIN bioreactor and finally scaled to two 15 L bioreactor. A final yield of 2.09 × 1010 cells with an overall expansion factor of 1975 within 13 days. The cells were harvested, concentrated, washed and prepared automatically with vivaPREP. The entire process was realized with ACISCP platform and was totally enclosed. Critical quality attributes (CQA) assessments and release tests of MSCs, including sterility, safety, purity, viability, identity, stability and potency were performed accordingly. The quality of cells harvested from 3D culture on the ACISCP and conventional 2D planar culture counterpart has no significant difference. This study provides a bioprocess engineering platform, harnessing GMP-grade 3D TableTrix® microcarriers and ACISCP to achieve industrial-scale manufacturing of clinical-grade hMSCs.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China.,Beijing CytoNiche Biotechnology Co. Ltd, Beijing, China
| | - Tao Na
- National Institutes for Food and Drug Control, Beijing, China
| | - Kehua Zhang
- National Institutes for Food and Drug Control, Beijing, China
| | - Yanping Yang
- Beijing CytoNiche Biotechnology Co. Ltd, Beijing, China
| | - Huanye Xu
- Beijing CytoNiche Biotechnology Co. Ltd, Beijing, China
| | - Lina Wei
- National Institutes for Food and Drug Control, Beijing, China
| | - Liming Xu
- National Institutes for Food and Drug Control, Beijing, China
| | - Xiaojun Yan
- Beijing CytoNiche Biotechnology Co. Ltd, Beijing, China
| | - Wei Liu
- Beijing CytoNiche Biotechnology Co. Ltd, Beijing, China
| | | | - Bin Wang
- Center for Clinic Stem Cell Research, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Shufang Meng
- National Institutes for Food and Drug Control, Beijing, China
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
31
|
Darge HF, Lin YH, Hsieh-Chih T, Lin SY, Yang MC. Thermo/redox-responsive dissolvable gelatin-based microsphere for efficient cell harvesting during 3D cell culturing. BIOMATERIALS ADVANCES 2022; 139:213008. [PMID: 35882154 DOI: 10.1016/j.bioadv.2022.213008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/21/2022] [Accepted: 06/28/2022] [Indexed: 06/15/2023]
Abstract
The use of microspheres for culturing adherent cells has been proven as an important method, allowing for obtaining adequate number of cells in limited space and volume of medium for the intended cell-based medical applications. However, the use of proteolytic enzymes for cell harvesting from the microsphere resulted in cell damage and loss of functionality. Therefore, in this study, we developed a novel redox/thermo-responsive dissolvable gelatin-based microsphere for successful cell proliferation and harvesting adequate high-quality cells using non-enzymatic cell detachment methods. Initially, a redox-induced dissolvable gelatin-based microsphere was successfully prepared using disulfide bonds as crosslinking agent, firmly stabilizing gelatin networks and forming a stable microsphere at physiological temperature. The optimized concentration of the crosslinking agent was 1.2 mM, which kept the microsphere stable for >120 h. The microsphere was then coated with PNIPAm-ALA copolymer via physical or chemical means, resulting in a positively charged thermosensitive surface. The positive charge derived from ALA in PNIPAm-ALA copolymer enhanced cell attachment, while the thermosensitive property of the copolymer enabled for temperature induced cell harvesting. When the temperature dropped below the LCST value of PNIPAm-ALA5 (33.4°C), the copolymer swelled and became more hydrophilic, allowing cells to be readily separated. The addition of reducing agents such as GSH, DTT and L-cysteine resulted in further cleavage of the disulfide bond in the microsphere and dissolution of the microsphere for complete cell detachment. Interestingly, cell attachment and proliferation were enhanced on microspheres coated with PNIPAm-ALA5 using diselenide as a crosslinking agent, and complete cell detachment was occurred within 15 min after adding 25 mM DTT followed by lowering the temperature (4°C). Therefore, the microsphere fabricated in this study was worthwhile for non-enzymatic cell detachment and has the potential to be used for cell expansion and harvesting adequate live cells of high quality and functionality for tissue engineering or cell therapy.
Collapse
Affiliation(s)
- Haile F Darge
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei, Taiwan; Advanced Membrane Materials Center, National Taiwan University of Science and Technology, Taipei, Taiwan; College of Medicine and Health Science, Bahir Dar University, Bahir Dar, Ethiopia
| | - Yu-Hsuan Lin
- Department of Materials Science and Engineering, National Taiwan University of Science and Technology, Taipei, Taiwan
| | - Tsai Hsieh-Chih
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei, Taiwan; Advanced Membrane Materials Center, National Taiwan University of Science and Technology, Taipei, Taiwan; R&D Center for Membrane Technology, Chung Yuan Christian University, Taoyuan, Taiwan.
| | - Shuian-Yin Lin
- Biomedical Technology and Device Research Center, Industrial Technology Research Institute, Hsinchu, Taiwan.
| | - Ming-Chien Yang
- Department of Materials Science and Engineering, National Taiwan University of Science and Technology, Taipei, Taiwan.
| |
Collapse
|
32
|
Liu Y, Wang R, Ding S, Deng L, Zhang Y, Li J, Shi Z, Wu Z, Liang K, Yan X, Liu W, Du Y. Engineered meatballs via scalable skeletal muscle cell expansion and modular micro-tissue assembly using porous gelatin micro-carriers. Biomaterials 2022; 287:121615. [PMID: 35679644 DOI: 10.1016/j.biomaterials.2022.121615] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 05/04/2022] [Accepted: 05/30/2022] [Indexed: 11/02/2022]
Abstract
The emerging field of cultured meat faces several technical hurdles, including the scale-up production of quality muscle and adipose progenitor cells, and the differentiation and bioengineering of these cellular materials into large, meat-like tissue. Here, we present edible, 3D porous gelatin micro-carriers (PoGelat-MCs), as efficient cell expansion scaffolds, as well as modular tissue-engineering building blocks for lab-grown meat. PoGelat-MC culture in spinner flasks, not only facilitated the scalable expansion of porcine skeletal muscle satellite cells and murine myoblasts, but also triggered their spontaneous myogenesis, in the absence of myogenic reagents. Using 3D-printed mold and transglutaminase, we bio-assembled pork muscle micro-tissues into centimeter-scale meatballs, which exhibited similar mechanical property and higher protein content compared to conventional ground pork meatballs. PoGelat-MCs also supported the expansion and differentiation of 3T3L1 murine pre-adipocytes into mature adipose micro-tissues, which could be used as modular assembly unit for engineered fat-containing meat products. Together, our results highlight PoGelat-MCs, in combination with dynamic bioreactors, as a scalable culture system to produce large quantity of highly-viable muscle and fat micro-tissues, which could be further bio-assembled into ground meat analogues.
Collapse
Affiliation(s)
- Ye Liu
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 10084, China
| | - Rui Wang
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 10084, China
| | - Shijie Ding
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, China
| | - Liping Deng
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 10084, China
| | - Yuanyuan Zhang
- Beijing CytoNiche Biotechnology Co. Ltd, Beijing, 100195, China
| | - Junyang Li
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 10084, China
| | - Ziao Shi
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 10084, China
| | - Zhongyuan Wu
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, China
| | - Kaini Liang
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 10084, China
| | - Xiaojun Yan
- Beijing CytoNiche Biotechnology Co. Ltd, Beijing, 100195, China
| | - Wei Liu
- Beijing CytoNiche Biotechnology Co. Ltd, Beijing, 100195, China
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 10084, China.
| |
Collapse
|
33
|
Liu X, Wang J, Xu X, Zhu H, Man K, Zhang J. SDF-1 Functionalized Hydrogel Microcarriers for Skin Flap Repair. ACS Biomater Sci Eng 2022; 8:3576-3588. [PMID: 35899941 DOI: 10.1021/acsbiomaterials.2c00755] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Critically sized skin flaps used to treat skin defects often suffer from necrosis due to insufficient blood supply. Hence there is an urgent need to improve the survival rate of skin flaps by promoting local angiogenesis. The delivery of growth factor loaded microcarriers have shown promise in enhancing defect repair, however, their rapid clearance from the defect site limits their regenerative potential. Thus, it is critical to develop microcarriers which can promote the sustained release of bioactive factors to effectively stimulate tissue repair. This study aimed to develop a stromal cell-derived factor 1 (SDF-1) loaded microcarrier coated with Matrigel (MC@SDF-1@Mat) to promote skin flap repair. SEM imaging showed that the surface of the microcarrier was coated by a porous Matrigel film. The drug release experiment showed that the Matrigel-coated microcarriers enhanced the sustained release of the model drug methylene blue when compared to uncoated group. MC@SDF-1@Mat significantly promoted the proliferation, migration, and angiogenesis of HUVECs via CCK-8, wound healing assay, and tube formation assay, respectively. Moreover, the murine random skin flap model was further established and treated. It was found that the flap necrosis area in the MC@SDF-1@Mat treated group was significantly reduced. H&E and Masson staining showed the histological structure and collagen organization exhibited a normal phenotype in the MC@SDF-1@Mat treated group. Additionally, CD31 immunohistochemical analysis showed that the MC@SDF-1@Mat treated group exhibited the greatest degree of neovascularization. In conclusion, our SDF-1 functionalized gelatin-based hydrogel microcarrier has potential clinical applications in promoting skin flap repair and drug delivery.
Collapse
Affiliation(s)
- Xiaochuan Liu
- Key Laboratory of 3D Printing Technology in Stomatology, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523710, P.R. China
| | - Jinsi Wang
- Key Laboratory of 3D Printing Technology in Stomatology, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523710, P.R. China
| | - Xiaoqin Xu
- Key Laboratory of 3D Printing Technology in Stomatology, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523710, P.R. China
| | - Hong Zhu
- Key Laboratory of 3D Printing Technology in Stomatology, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523710, P.R. China
| | - Kenny Man
- School of Chemical Engineering, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Jingying Zhang
- Key Laboratory of 3D Printing Technology in Stomatology, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523710, P.R. China
| |
Collapse
|
34
|
Feng ZY, Zhang QY, Tan J, Xie HQ. Techniques for increasing the yield of stem cell-derived exosomes: what factors may be involved? SCIENCE CHINA. LIFE SCIENCES 2022; 65:1325-1341. [PMID: 34637101 PMCID: PMC8506103 DOI: 10.1007/s11427-021-1997-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 08/11/2021] [Indexed: 02/05/2023]
Abstract
Exosomes are nano-scale extracellular vesicles secreted by cells and constitute an important part in the cell-cell communication. The main contents of the exosomes include proteins, microRNAs, and lipids. The mechanism and safety of stem cell-derived exosomes have rendered them a promising therapeutic strategy for regenerative medicine. Nevertheless, limited yield has restrained full explication of their functions and clinical applications To address this, various attempts have been made to explore the up- and down-stream manipulations in a bid to increase the production of exosomes. This review has recapitulated factors which may influence the yield of stem cell-derived exosomes, including selection and culture of stem cells, isolation and preservation of the exosomes, and development of artificial exosomes.
Collapse
Affiliation(s)
- Zi-Yuan Feng
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Med-X Center for Materials, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qing-Yi Zhang
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Med-X Center for Materials, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jie Tan
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Med-X Center for Materials, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hui-Qi Xie
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Med-X Center for Materials, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
35
|
Timsina H, McTyer J, Rao RR, Almodovar J. A comparative evaluation of layer-by-layer assembly techniques for surface modification of microcarriers used in human mesenchymal stromal cell manufacturing. Biotechnol J 2022; 17:e2100605. [PMID: 35377534 DOI: 10.1002/biot.202100605] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 03/16/2022] [Accepted: 04/01/2022] [Indexed: 11/06/2022]
Abstract
The demand for large quantities of highly potent human mesenchymal stromal cells (hMSCs) is growing given their therapeutic potential. To meet high production needs, suspension-based cell cultures using microcarriers are commonly used. Microcarriers are commonly made of or coated with extracellular matrix proteins or charged compounds to promote cell adhesion and proliferation. In this work, we demonstrate a simple method (draining filter) to perform layer by layer (LbL) assembly on microcarriers to create multilayers of heparin and collagen and further demonstrate that these multilayers have a positive effect on hMSC viability after 48 hours of culture. The draining filter method is evaluated against two other methods found in literature - centrifugation and fluidized bed, showing that the draining filter method can perform the surface modification with greater efficiency and with less materials and steps needed in the coating process. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Hemanta Timsina
- Ralph E. Martin Department of Chemical Engineering, University of Arkansas, 3202 Bell Engineering Center, Fayetteville, AR, 72701, USA
| | - Jasmine McTyer
- Department of Chemical and Biomolecular Engineering, Clemson University, 127 Earle Hall, Clemson, SC, 29634, USA
| | - Raj R Rao
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, 72701, USA
| | - Jorge Almodovar
- Ralph E. Martin Department of Chemical Engineering, University of Arkansas, 3202 Bell Engineering Center, Fayetteville, AR, 72701, USA
| |
Collapse
|
36
|
Functional tissue-engineered microtissue formed by self-aggregation of cells for peripheral nerve regeneration. Stem Cell Res Ther 2022; 13:3. [PMID: 35012663 PMCID: PMC8744299 DOI: 10.1186/s13287-021-02676-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 12/11/2021] [Indexed: 02/06/2023] Open
Abstract
Background Peripheral nerve injury (PNI) is one of the essential causes of physical disability with a high incidence rate. The traditional tissue engineering strategy, Top-Down strategy, has some limitations. A new tissue-engineered strategy, Bottom-Up strategy (tissue-engineered microtissue strategy), has emerged and made significant research progress in recent years. However, to the best of our knowledge, microtissues are rarely used in neural tissue engineering; thus, we intended to use microtissues to repair PNI.
Methods We used a low-adhesion cell culture plate to construct adipose-derived mesenchymal stem cells (ASCs) into microtissues in vitro, explored the physicochemical properties and microtissues components, compared the expression of cytokines related to nerve regeneration between microtissues and the same amount of two-dimension (2D)-cultured cells, co-cultured directly microtissues with dorsal root ganglion (DRG) or Schwann cells (SCs) to observe the interaction between them using immunocytochemistry, quantitative reverse transcription polymerase chain reaction (qRT-PCR), enzyme-linked immunosorbent assay (ELISA). We used grafts constructed by microtissues and polycaprolactone (PCL) nerve conduit to repair sciatic nerve defects in rats. Results The present study results indicated that compared with the same number of 2D-cultured cells, microtissue could secrete more nerve regeneration related cytokines to promote SCs proliferation and axons growth. Moreover, in the direct co-culture system of microtissue and DRG or SCs, axons of DRG grown in the direction of microtissue, and there seems to be a cytoplasmic exchange between SCs and ASCs around microtissue. Furthermore, microtissues could repair sciatic nerve defects in rat models more effectively than traditional 2D-cultured ASCs. Conclusion Tissue-engineered microtissue is an effective strategy for stem cell culture and therapy in nerve tissue engineering. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02676-0.
Collapse
|
37
|
Wang B, Liu W, Li JJ, Chai S, Xing D, Yu H, Zhang Y, Yan W, Xu Z, Zhao B, Du Y, Jiang Q. A low dose cell therapy system for treating osteoarthritis: In vivo study and in vitro mechanistic investigations. Bioact Mater 2022; 7:478-490. [PMID: 34466747 PMCID: PMC8379370 DOI: 10.1016/j.bioactmat.2021.05.029] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 04/28/2021] [Accepted: 05/17/2021] [Indexed: 12/17/2022] Open
Abstract
Mesenchymal stem cells (MSCs) can be effective in alleviating the progression of osteoarthritis (OA). However, low MSC retention and survival at the injection site frequently require high doses of cells and/or repeated injections, which are not economically viable and create additional risks of complications. In this study, we produced MSC-laden microcarriers in spinner flask culture as cell delivery vehicles. These microcarriers containing a low initial dose of MSCs administered through a single injection in a rat anterior cruciate ligament (ACL) transection model of OA achieved similar reparative effects as repeated high doses of MSCs, as evaluated through imaging and histological analyses. Mechanistic investigations were conducted using a co-culture model involving human primary chondrocytes grown in monolayer, together with MSCs grown either within 3D constructs or as a monolayer. Co-culture supernatants subjected to secretome analysis showed significant decrease of inflammatory factors in the 3D group. RNA-seq of co-cultured MSCs and chondrocytes using Gene Ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis revealed processes relating to early chondrogenesis and increased extracellular matrix interactions in MSCs of the 3D group, as well as phenotypic maintenance in the co-cultured chondrocytes. The cell delivery platform we investigated may be effective in reducing the cell dose and injection frequency required for therapeutic applications.
Collapse
Affiliation(s)
- Bin Wang
- Department of Sports Medicine and Adult Reconstruction Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 201180, China
- Department of Orthopaedics, Shanxi Medical University Second Affiliated Hospital, Taiyuan, 030001, China
| | - Wei Liu
- Beijing CytoNiche Biotechnology Co. Ltd, Beijing, 10081, China
| | - Jiao Jiao Li
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Senlin Chai
- Department of Sports Medicine and Adult Reconstruction Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 201180, China
- Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing, 210093, China
| | - Dan Xing
- Arthritis Clinic & Research Center, Peking University People's Hospital, Peking University, Beijing, 100044, China
| | - Hongsheng Yu
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yuanyuan Zhang
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Wenjin Yan
- Department of Sports Medicine and Adult Reconstruction Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 201180, China
- Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing, 210093, China
| | - Zhihong Xu
- Department of Sports Medicine and Adult Reconstruction Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 201180, China
- Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing, 210093, China
| | - Bin Zhao
- Department of Orthopaedics, Shanxi Medical University Second Affiliated Hospital, Taiyuan, 030001, China
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Qing Jiang
- Department of Sports Medicine and Adult Reconstruction Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 201180, China
- Laboratory for Bone and Joint Disease, Model Animal Research Center (MARC), Nanjing University, Nanjing, 210093, China
| |
Collapse
|
38
|
Chen J, Zhou D, Nie Z, Lu L, Lin Z, Zhou D, Zhang Y, Long X, Fan S, Xu T. A scalable coaxial bioprinting technology for mesenchymal stem cell microfiber fabrication and high extracellular vesicle yield. Biofabrication 2021; 14:015012. [PMID: 34798619 DOI: 10.1088/1758-5090/ac3b90] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 11/19/2021] [Indexed: 02/08/2023]
Abstract
Mesenchymal stem cell (MSC)-derived extracellular vesicles (EVs) are promising candidates for regenerative medicine; however, the lack of scalable methods for high quantity EV production limits their application. In addition, signature EV-derived proteins shared in 3D environments and 2D surfaces, remain mostly unknown. Herein, we present a platform combining MSC microfiber culture with ultracentrifugation purification for high EV yield. Within this platform, a high quantity MSC solution (∼3 × 108total cells) is encapsulated in a meter-long hollow hydrogel-microfiber via coaxial bioprinting technology. In this 3D core-shell microfiber environment, MSCs express higher levels of stemness markers (Oct4, Nanog, Sox2) than in 2D culture, and maintain their differentiation capacity. Moreover, this platform enriches particles by ∼1009-fold compared to conventional 2D culture, while preserving their pro-angiogenic properties. Liquid chromatography-mass spectrometry characterization results demonstrate that EVs derived from our platform and conventional 2D culturing have unique protein profiles with 3D-EVs having a greater variety of proteins (1023 vs 605), however, they also share certain proteins (536) and signature MSC-EV proteins (10). This platform, therefore, provides a new tool for EV production using microfibers in one culture dish, thereby reducing space, labor, time, and cost.
Collapse
Affiliation(s)
- Jianwei Chen
- Precision Medicine and Healthcare Research Center, Tsinghua-Berkeley Shenzhen Institute (TBSI), Tsinghua University, Shenzhen 518055, People's Republic of China
| | - Duchao Zhou
- East China Institute of Digital Medical Engineering, Shangrao 334000, People's Republic of China
| | - Zhenguo Nie
- Department of Orthopedics, Fourth Medical Center of PLA general hospital, 100048 Beijing, People's Republic of China
| | - Liang Lu
- Department of Neurosurgery, The First Affiliated Hospital of Shantou University Medical College, 515041 Shantou, Guangdong, People's Republic of China
| | - Zhidong Lin
- The Second Affiliated Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, 510006 Guangzhou, People's Republic of China
| | - Dezhi Zhou
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China
| | - Yi Zhang
- Precision Medicine and Healthcare Research Center, Tsinghua-Berkeley Shenzhen Institute (TBSI), Tsinghua University, Shenzhen 518055, People's Republic of China
| | - Xiaoyan Long
- East China Institute of Digital Medical Engineering, Shangrao 334000, People's Republic of China
| | - Siyang Fan
- Heart Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, People's Republic of China
| | - Tao Xu
- Precision Medicine and Healthcare Research Center, Tsinghua-Berkeley Shenzhen Institute (TBSI), Tsinghua University, Shenzhen 518055, People's Republic of China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China
- Key Laboratory for Advanced Materials Processing Technology, Ministry of Education, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China
| |
Collapse
|
39
|
Xu H, Cong Z, Zhang Y, Liu W, Yan X, Du Y. Large-Scale Expansion of Umbilical Cord Mesenchymal Stem Cells with Microcarrier Tablets in Bioreactor. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2021; 2436:113-125. [PMID: 34519979 DOI: 10.1007/7651_2021_425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Mesenchymal stem cells show great potential in tissue engineering and cell-based therapies. This protocol demonstrates the use of 3D TableTrix® microcarrier tablets for large-scale manufacturing of human umbilical cord mesenchymal stem cells (hUCMSCs) in a 5-L stirred tank bioreactor. 3D TableTrix® microcarrier tablets readily disperse into tens of thousands of porous microcarriers to simplify cell seeding, and 3D FloTrix® vivaSPIN bioreactor could automate, monitor, and control the entire culture process. 3D TableTrix® microcarriers could also be fully dissolved upon adding dissolution reagent to gently harvest the expanded cells at a high recovery rate. With this protocol, more than 109 cells could be produced in a 5-L bioreactor.
Collapse
Affiliation(s)
- Huanye Xu
- Beijing CytoNiche Biotechnology Co., Ltd., Beijing, China
| | - Zhongxiao Cong
- Beijing CytoNiche Biotechnology Co., Ltd., Beijing, China
| | - Yuanyuan Zhang
- Beijing CytoNiche Biotechnology Co., Ltd., Beijing, China
| | - Wei Liu
- Beijing CytoNiche Biotechnology Co., Ltd., Beijing, China
| | - Xiaojun Yan
- Beijing CytoNiche Biotechnology Co., Ltd., Beijing, China.
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Tsinghua-PKU Center for Life Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
40
|
Tsai AC, Pacak CA. Bioprocessing of Human Mesenchymal Stem Cells: From Planar Culture to Microcarrier-Based Bioreactors. Bioengineering (Basel) 2021; 8:bioengineering8070096. [PMID: 34356203 PMCID: PMC8301102 DOI: 10.3390/bioengineering8070096] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 07/02/2021] [Accepted: 07/02/2021] [Indexed: 01/14/2023] Open
Abstract
Human mesenchymal stem cells (hMSCs) have demonstrated great potential to be used as therapies for many types of diseases. Due to their immunoprivileged status, allogeneic hMSCs therapies are particularly attractive options and methodologies to improve their scaling and manufacturing are needed. Microcarrier-based bioreactor systems provide higher volumetric hMSC production in automated closed systems than conventional planar cultures. However, more sophisticated bioprocesses are necessary to successfully convert from planar culture to microcarriers. This article summarizes key steps involved in the planar culture to microcarrier hMSC manufacturing scheme, from seed train, inoculation, expansion and harvest. Important bioreactor parameters, such as temperature, pH, dissolved oxygen (DO), mixing, feeding strategies and cell counting techniques, are also discussed.
Collapse
Affiliation(s)
- Ang-Chen Tsai
- Department of Pediatrics, University of Florida, Gainesville, FL 32603, USA
- Correspondence: (A.-C.T.); (C.A.P.)
| | - Christina A. Pacak
- Department of Neurology, University of Minnesota, Minneapolis, MN 55455, USA
- Correspondence: (A.-C.T.); (C.A.P.)
| |
Collapse
|
41
|
Zhang Y, Gao S, Liang K, Wu Z, Yan X, Liu W, Li J, Wu B, Du Y. Exendin-4 gene modification and microscaffold encapsulation promote self-persistence and antidiabetic activity of MSCs. SCIENCE ADVANCES 2021; 7:eabi4379. [PMID: 34215590 PMCID: PMC11060038 DOI: 10.1126/sciadv.abi4379] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 05/20/2021] [Indexed: 06/13/2023]
Abstract
Mesenchymal stem cell (MSC)-based therapy to combat diabetic-associated metabolic disorders is hindered by impoverished cell survival and limited therapeutic effects under high glucose stress. Here, we genetically engineered MSCs with Exendin-4 (MSC-Ex-4), a glucagon-like peptide-1 (GLP-1) analog, and demonstrated their boosted cellular functions and antidiabetic efficacy in the type 2 diabetes mellitus (T2DM) mouse model. Mechanistically, MSC-Ex-4 achieved self-augmentation and improved survival under high glucose stress via autocrine activation of the GLP-1R-mediated AMPK signaling pathway. Meanwhile, MSC-Ex-4-secreted Exendin-4 suppressed senescence and apoptosis of pancreatic β cells through endocrine effects, while MSC-Ex-4-secreted bioactive factors (e.g., IGFBP2 and APOM) paracrinely augmented insulin sensitivity and decreased lipid accumulation in hepatocytes through PI3K-Akt activation. Furthermore, we encapsulated MSC-Ex-4 in 3D gelatin microscaffolds for single-dose administration to extend the therapeutic effect for 3 months. Together, our findings provide mechanistic insights into Exendin-4-mediated MSCs self-persistence and antidiabetic activity that offer more effective MSC-based therapy for T2DM.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Shuang Gao
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Kaini Liang
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Zhaozhao Wu
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xiaojun Yan
- Beijing CytoNiche Biotechnology Co. Ltd., Beijing 100195, China
| | - Wei Liu
- Beijing CytoNiche Biotechnology Co. Ltd., Beijing 100195, China
| | - Junyang Li
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Bingjie Wu
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
42
|
Zhang J, Xu W, Li C, Meng F, Guan Y, Liu X, Zhao J, Peng J, Wang Y. Tissue Engineering Microtissue: Construction, Optimization, and Application. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:393-404. [PMID: 33719547 DOI: 10.1089/ten.teb.2020.0370] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Until now, there is no clear definition of microtissue; it usually refers to the microtissue formed by the aggregation of seed cells under the action of cell-cell or cell-extracellular matrix (ECM). Compared with traditional cell monolayer culture, cells are cultivated into a three-dimensional microstructure in a specific way. The microstructure characteristics of microtissue are similar to natural tissues and can promote cell proliferation and differentiation. Therefore, it has a broader range of biomedical applications in tissue engineering. The traditional tissue engineering strategy is to add high-density seed cells and biomolecules on a preformed scaffold to construct a tissue engineering graft. However, due to the destruction of the ECM of the cells cultured in a monolayer during the digestion process with trypsin, the uneven distribution of the cells in the scaffold, and the damage of various adverse factors after the cells are implanted in the scaffold, this strategy is often ineffective, and the subsequent applications still face challenges. This article reviews the latest researches of a new strategy-tissue engineering microtissue strategy; discuss several traditional construction methods, structure, and function optimization; and practical application of microtissue. The review aims to provide a reference for future research on tissue engineering microtissue. Impact statement The traditional tissue engineering strategies have several disadvantages, researchers have conducted extensive research on tissue engineering microtissues in recent years, and they make significant progress. Microtissue is a kind of microtissue with three-dimensional structure, its microstructure is similar to that of natural tissue. In addition, microtissue implantation can protect cells from mechanical interference, inflammation, and other adverse factors. Furthermore, it improves the survival rate of cells and the therapeutic effect of tissue-engineered grafts. However, the practical conditions, advantages, and disadvantages of tissue engineering microtissue have not been fully elucidated. The purpose of this review is to discuss the latest research progress of microtissue and provide a reference for future research.
Collapse
Affiliation(s)
- Jian Zhang
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, P.R. China
| | - Wenjing Xu
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, P.R. China
| | - Chaochao Li
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, P.R. China
| | - Fanqi Meng
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, P.R. China.,Department of Spine Surgery, Peking University People's Hospital, Beijing, P.R. China
| | - Yanjun Guan
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, P.R. China
| | - Xiuzhi Liu
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, P.R. China
| | - Jie Zhao
- Beijing Tsinghua Changgeng Hospital Affiliated to Tsinghua University, Tsinghua University Clinical School, Beijing, P.R. China
| | - Jiang Peng
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, P.R. China.,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, P.R. China
| | - Yu Wang
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institute of Orthopedics, Chinese PLA General Hospital, Beijing, P.R. China.,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, P.R. China
| |
Collapse
|
43
|
Muoio F, Panella S, Jossen V, Lindner M, Harder Y, Müller M, Eibl R, Tallone T. Human Adipose Stem Cells (hASCs) Grown on Biodegradable Microcarriers in Serum- and Xeno-Free Medium Preserve Their Undifferentiated Status. J Funct Biomater 2021; 12:jfb12020025. [PMID: 33923488 PMCID: PMC8167760 DOI: 10.3390/jfb12020025] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/25/2021] [Accepted: 04/12/2021] [Indexed: 12/17/2022] Open
Abstract
Human adipose stem cells (hASCs) are promising candidates for cell-based therapies, but they need to be efficiently expanded in vitro as they cannot be harvested in sufficient quantities. Recently, dynamic bioreactor systems operated with microcarriers achieved considerable high cell densities. Thus, they are a viable alternative to static planar cultivation systems to obtain high numbers of clinical-grade hASCs. Nevertheless, the production of considerable biomass in a short time must not be achieved to the detriment of the cells' quality. To facilitate the scalable expansion of hASC, we have developed a new serum- and xeno-free medium (UrSuppe) and a biodegradable microcarrier (BR44). In this study, we investigated whether the culture of hASCs in defined serum-free conditions on microcarriers (3D) or on planar (2D) cell culture vessels may influence the expression of some marker genes linked with the immature degree or the differentiated status of the cells. Furthermore, we investigated whether the biomaterials, which form our biodegradable MCs, may affect cell behavior and differentiation. The results confirmed that the quality and the undifferentiated status of the hASCs are very well preserved when they grow on BR44 MCs in defined serum-free conditions. Indeed, the ASCs showed a gene expression profile more compatible with an undifferentiated status than the same cells grown under standard planar conditions.
Collapse
Affiliation(s)
- Francesco Muoio
- Foundation for Cardiological Research and Education (FCRE), Cardiocentro Ticino Foundation, 6807 Taverne, Switzerland; (F.M.); (S.P.)
| | - Stefano Panella
- Foundation for Cardiological Research and Education (FCRE), Cardiocentro Ticino Foundation, 6807 Taverne, Switzerland; (F.M.); (S.P.)
| | - Valentin Jossen
- Institute of Chemistry & Biotechnology, Competence Center of Biochemical Engineering & Cell Cultivation Technique Zurich University of Applied Sciences, 8820 Wädenswil, Switzerland; (V.J.); (R.E.)
| | | | - Yves Harder
- Department of Plastic, Reconstructive and Aesthetic Surgery, EOC, 6900 Lugano, Switzerland;
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6900 Lugano, Switzerland
| | | | - Regine Eibl
- Institute of Chemistry & Biotechnology, Competence Center of Biochemical Engineering & Cell Cultivation Technique Zurich University of Applied Sciences, 8820 Wädenswil, Switzerland; (V.J.); (R.E.)
| | - Tiziano Tallone
- Foundation for Cardiological Research and Education (FCRE), Cardiocentro Ticino Foundation, 6807 Taverne, Switzerland; (F.M.); (S.P.)
- Correspondence: ; Tel.: +41-91-805-38-85
| |
Collapse
|
44
|
Yu H, You Z, Yan X, Liu W, Nan Z, Xing D, Huang C, Du Y. TGase-Enhanced Microtissue Assembly in 3D-Printed-Template-Scaffold (3D-MAPS) for Large Tissue Defect Reparation. Adv Healthc Mater 2020; 9:e2000531. [PMID: 32803857 DOI: 10.1002/adhm.202000531] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 06/30/2020] [Indexed: 01/08/2023]
Abstract
Engineering large and functional tissue constructs with complex structures (e.g., external ear or nose) for reparation and reconstruction of tissue defects remains one of the major challenges in regenerative medicine, which demands abundant cell sources, advanced biofabrication schemes, and satisfactory integration with the host for long-term efficacy post implantation. Here the 'Microtissue Assembly in 3D-Printed-template-Scaffold' (3D-MAPS), as a platform technology to rapidly fabricate centimeter-sized functional tissue constructs with complex structures, is developed. 3D-MAPS facilitates bottom-up assembly of large-scale manufactured microtissues within the 3D-printed hollow polymeric templates with pre-defined architectures. The assembly and fusion of 2×106 mesenchymal stem cell-based microtissues within the defined 3D-printed template is further enhanced by addition of a natural protein crosslinker (i.e., transglutaminase (TGase)), and thereby achieves construction of centimeter-sized tissue with high cell viability and mechanical stability in vitro within 30 min. Further in vivo implantation of the 3D-MAPS-fabricated ear-like tissue construct in rabbit models assisted by flap prefabrication technique results in increased structural vascular support and strengthened functional survival. Thus, the TGase-enhanced 3D-MAPS demonstrates its potential and feasibility as a powerful biofabrication platform for tissue engineering application.
Collapse
Affiliation(s)
- Hongsheng Yu
- Department of Biomedical Engineering, School of Medicine Tsinghua‐Peking Center for Life Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology Tsinghua University Beijing 100084 China
| | - Zhifeng You
- Department of Biomedical Engineering, School of Medicine Tsinghua‐Peking Center for Life Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology Tsinghua University Beijing 100084 China
| | - Xiaojun Yan
- Beijing CytoNiche Biotechnology Co. Ltd. Beijing 100195 China
| | - Wei Liu
- Beijing CytoNiche Biotechnology Co. Ltd. Beijing 100195 China
| | - Zhirui Nan
- Department of Biomedical Engineering, School of Medicine Tsinghua‐Peking Center for Life Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology Tsinghua University Beijing 100084 China
| | - Dan Xing
- Arthritis Clinic and Research Center, Peking University People's Hospital Peking University Beijing 100044 China
| | - Chenyu Huang
- Department of Dermatology Beijing Tsinghua Changgung Hospital School of Clinical Medicine Tsinghua University Beijing 102218 China
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine Tsinghua‐Peking Center for Life Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology Tsinghua University Beijing 100084 China
| |
Collapse
|
45
|
Tsai AC, Jeske R, Chen X, Yuan X, Li Y. Influence of Microenvironment on Mesenchymal Stem Cell Therapeutic Potency: From Planar Culture to Microcarriers. Front Bioeng Biotechnol 2020; 8:640. [PMID: 32671039 PMCID: PMC7327111 DOI: 10.3389/fbioe.2020.00640] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 05/26/2020] [Indexed: 12/15/2022] Open
Abstract
Human mesenchymal stem cells (hMSCs) are a promising candidate in cell therapy as they exhibit multilineage differentiation, homing to the site of injury, and secretion of trophic factors that facilitate tissue healing and/or modulate immune response. As a result, hMSC-derived products have attracted growing interests in preclinical and clinical studies. The development of hMSC culture platforms for large-scale biomanufacturing is necessary to meet the requirements for late-phase clinical trials and future commercialization. Microcarriers in stirred-tank bioreactors have been widely utilized in large-scale expansion of hMSCs for translational applications because of a high surface-to-volume ratio compared to conventional 2D planar culture. However, recent studies have demonstrated that microcarrier-expanded hMSCs differ from dish- or flask-expanded cells in size, morphology, proliferation, viability, surface markers, gene expression, differentiation potential, and secretome profile which may lead to altered therapeutic potency. Therefore, understanding the bioprocessing parameters that influence hMSC therapeutic efficacy is essential for the optimization of microcarrier-based bioreactor system to maximize hMSC quantity without sacrificing quality. In this review, biomanufacturing parameters encountered in planar culture and microcarrier-based bioreactor culture of hMSCs are compared and discussed with specific focus on cell-adhesion surface (e.g., discontinuous surface, underlying curvature, microcarrier stiffness, porosity, surface roughness, coating, and charge) and the dynamic microenvironment in bioreactor culture (e.g., oxygen and nutrients, shear stress, particle collision, and aggregation). The influence of dynamic culture in bioreactors on hMSC properties is also reviewed in order to establish connection between bioprocessing and stem cell function. This review addresses fundamental principles and concepts for future design of biomanufacturing systems for hMSC-based therapy.
Collapse
Affiliation(s)
- Ang-Chen Tsai
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, United States
| | - Richard Jeske
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, United States
| | - Xingchi Chen
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, United States
| | - Xuegang Yuan
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, United States
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, United States
| |
Collapse
|