1
|
Ralph PC, Choi SW, Baek MJ, Lee SJ. Regenerative medicine approaches for the treatment of spinal cord injuries: Progress and challenges. Acta Biomater 2024:S1742-7061(24)00613-5. [PMID: 39424019 DOI: 10.1016/j.actbio.2024.10.021] [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: 07/01/2024] [Revised: 10/03/2024] [Accepted: 10/15/2024] [Indexed: 10/21/2024]
Abstract
Spinal cord injury (SCI) is a profound medical condition that significantly hampers motor function, imposing substantial limitations on daily activities and exerting a considerable financial burden on patients and their families. The constrained regenerative capacity of endogenous spinal cord tissue, exacerbated by the inflammatory response following the initial trauma, poses a formidable obstacle to effective therapy. Recent advancements in the field, stem cells, biomaterials, and molecular therapy, show promising outcomes. This review provides a comprehensive analysis of tissue engineering and regenerative medicine approaches for SCI treatment, including cell transplantation, tissue-engineered construct implantation, and other potential therapeutic strategies. Additionally, it sheds light on preclinical animal studies and recent clinical trials incorporating these modalities, providing a glimpse into the evolving landscape of SCI management. STATEMENT OF SIGNIFICANCE: The investigation into spinal cord injury (SCI) treatments focuses on reducing long-term impacts by targeting scar inhibition and enhancing regeneration through stem cells, with or without growth factors. Induced pluripotent stem cells (iPSCs) show promise for autologous use, with clinical trials confirming their safety. Challenges include low cell viability and difficulty in targeted differentiation. Biomaterial scaffolds hold potential for improving cell viability and integration, and extracellular vesicles (EVs) are emerging as a novel therapy. While EV research is in its early stages, stem cell trials demonstrate safety and potential recovery. Advancing tissue engineering approaches with biomaterial scaffolds is crucial for human trials.
Collapse
Affiliation(s)
- Patrick C Ralph
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157, United States
| | - Sung-Woo Choi
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157, United States; Department of Orthopedic Surgery, Soonchunhyang University Hospital Seoul, Seoul 04401, Republic of Korea
| | - Min Jung Baek
- Department of Obstetrics and Gynecology, CHA University Bundang Medical Center, Seongnam, Gyeonggi-do 13496, Republic of Korea
| | - Sang Jin Lee
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157, United States.
| |
Collapse
|
2
|
Dadvand A, Yavari A, Teimourpour A, Farzad-Mohajeri S. Influential factors on stem cell therapy success in canine model of spinal cord Injury: A systematic review and meta-analysis. Brain Res 2024; 1839:148997. [PMID: 38795792 DOI: 10.1016/j.brainres.2024.148997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 05/03/2024] [Accepted: 05/10/2024] [Indexed: 05/28/2024]
Abstract
Spinal cord injury (SCI) is a serious medical condition. The search for an effective cure remains a persistent challenge. Current treatments, unfortunately, are unable to sufficiently improve neurological function, often leading to lifelong disability. This systematic review and meta-analysis evaluated the effectiveness of stem cell therapy for SCI using canine models. It also explored the optimal protocol for implementing stem cell therapy. A comprehensive search of studies was conducted from 2000 to October 2022. This study focused on five outcomes: motor function score, histopathology, IHC, western blot, and SEP. The results demonstrated a significant improvement in locomotion post-SCI in dogs treated with stem cell therapy. The therapy also led to an average increase of 3.15 points in the Olby score of the treated dogs compared to the control group. These findings highlights stem cell therapy's potential as a promising SCI treatment. The meta-analysis suggests that using bone marrow stem cells, undergoing neural differentiation in vitro, applying a surgical implantation or intrathecal route of administration, associating matrigel in combination with stem cells, and a waiting period of two weeks before starting treatment can enhance SCI treatment effectiveness.
Collapse
Affiliation(s)
- Avin Dadvand
- Department of Surgery and Radiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| | - Alimohammad Yavari
- Department of Surgery and Radiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| | - Amir Teimourpour
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Saeed Farzad-Mohajeri
- Department of Surgery and Radiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran; Department of Regenerative Medicine, Institute of Biomedical Research, University of Tehran, Tehran, Iran.
| |
Collapse
|
3
|
Dallatana A, Cremonesi L, Pezzini F, Fontana G, Innamorati G, Giacomello L. The Placenta as a Source of Human Material for Neuronal Repair. Biomedicines 2024; 12:1567. [PMID: 39062139 PMCID: PMC11275125 DOI: 10.3390/biomedicines12071567] [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: 06/04/2024] [Revised: 07/08/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Stem cell therapy has the potential to meet unsolved problems in tissue repair and regeneration, particularly in the neural tissues. However, an optimal source has not yet been found. Growing evidence indicates that positive effects produced in vivo by mesenchymal stem cells (MSCs) can be due not only to their plasticity but also to secreted molecules including extracellular vesicles (EVs) and the extracellular matrix (ECM). Trophic effects produced by MSCs may reveal the key to developing effective tissue-repair strategies, including approaches based on brain implants or other implantable neural electrodes. In this sense, MSCs will become increasingly valuable and needed in the future. The placenta is a temporary organ devoted to protecting and supporting the fetus. At the same time, the placenta represents an abundant and extremely convenient source of MSCs. Nonetheless, placenta-derived MSCs (P-MSCs) remain understudied as compared to MSCs isolated from other sources. This review outlines the limited literature describing the neuroregenerative effects of P-MSC-derived biomaterials and advocates for exploiting the potential of this untapped source for human regenerative therapies.
Collapse
Affiliation(s)
| | | | | | | | - Giulio Innamorati
- Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, University of Verona, 37134 Verona, Italy; (A.D.); (L.C.); (F.P.); (G.F.); (L.G.)
| | | |
Collapse
|
4
|
Tan Z, Xiao L, Ma J, Shi K, Liu J, Feng F, Xie P, Dai Y, Yuan Q, Wu W, Rong L, He L. Integrating hydrogels manipulate ECM deposition after spinal cord injury for specific neural reconnections via neuronal relays. SCIENCE ADVANCES 2024; 10:eado9120. [PMID: 38959311 PMCID: PMC11221524 DOI: 10.1126/sciadv.ado9120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 05/30/2024] [Indexed: 07/05/2024]
Abstract
A bioinspired hydrogel composed of hyaluronic acid-graft-dopamine (HADA) and a designer peptide HGF-(RADA)4-DGDRGDS (HRR) was presented to enhance tissue integration following spinal cord injury (SCI). The HADA/HRR hydrogel manipulated the infiltration of PDGFRβ+ cells in a parallel pattern, transforming dense scars into an aligned fibrous substrate that guided axonal regrowth. Further incorporation of NT3 and curcumin promoted axonal regrowth and survival of interneurons at lesion borders, which served as relays for establishing heterogeneous axon connections in a target-specific manner. Notable improvements in motor, sensory, and bladder functions resulted in rats with complete spinal cord transection. The HADA/HRR + NT3/Cur hydrogel promoted V2a neuron accumulation in ventral spinal cord, facilitating the recovery of locomotor function. Meanwhile, the establishment of heterogeneous neural connections across the hemisected lesion of canines was documented in a target-specific manner via neuronal relays, significantly improving motor functions. Therefore, biomaterials can inspire beneficial biological activities for SCI repair.
Collapse
Affiliation(s)
- Zan Tan
- Department of Spine Surgery, The 3rd Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Longyou Xiao
- Department of Spine Surgery, The 3rd Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Junwu Ma
- Department of Spine Surgery, The 3rd Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Kaixi Shi
- Department of Spine Surgery, The 3rd Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Jialin Liu
- Department of Spine Surgery, The 3rd Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Feng Feng
- Department of Spine Surgery, The 3rd Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Pengfei Xie
- Department of Spine Surgery, The 3rd Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Yu Dai
- Department of Spine Surgery, The 3rd Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Qiuju Yuan
- Centre of Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong 999077, China
| | - Wutian Wu
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou 510632, China
- Re-Stem Biotechnology Co. Ltd., Suzhou 215129, China
| | - Limin Rong
- Department of Spine Surgery, The 3rd Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Liumin He
- Department of Spine Surgery, The 3rd Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| |
Collapse
|
5
|
Liu J, Yan R, Wang B, Chen S, Hong H, Liu C, Chen X. Decellularized extracellular matrix enriched with GDNF enhances neurogenesis and remyelination for improved motor recovery after spinal cord injury. Acta Biomater 2024; 180:308-322. [PMID: 38615813 DOI: 10.1016/j.actbio.2024.04.015] [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: 10/07/2023] [Revised: 04/08/2024] [Accepted: 04/09/2024] [Indexed: 04/16/2024]
Abstract
Motor functional improvement represents a paramount treatment objective in the post-spinal cord injury (SCI) recovery process. However, neuronal cell death and axonal degeneration following SCI disrupt neural signaling, impeding the motor functional recovery. In this study, we developed a multifunctional decellularized spinal cord-derived extracellular matrix (dSECM), crosslinked with glial cell-derived neurotrophic factor (GDNF), to promote differentiation of stem cells into neural-like cells and facilitate axonogenesis and remyelination. After decellularization, the immunogenic cellular components were effectively removed in dSECM, while the crucial protein components were retained which supports stem cells proliferation and differentiation. Furthermore, sustained release of GDNF from the dSECM facilitated axonogenesis and remyelination by activating the PI3K/Akt and MEK/Erk pathways. Our findings demonstrate that the dSECM-GDNF platform promotes neurogenesis, axonogenesis, and remyelination to enhance neural signaling, thereby yielding promising therapeutic effects for motor functional improvement after SCI. STATEMENT OF SIGNIFICANCE: The dSECM promotes the proliferation and differentiation of MSCs or NSCs by retaining proteins associated with positive regulation of neurogenesis and neuronal differentiation, while eliminating proteins related to negative regulation of neurogenesis. After crosslinking, GDNF can be gradually released from the platform, thereby promoting neural differentiation, axonogenesis, and remyelination to enhance neural signaling through activation of the PI3K/Akt and MEK/Erk pathways. In vivo experiments demonstrated that dSECM-GDNF/MSC@GelMA hydrogel exhibited the ability to facilitate neuronal regeneration at 4 weeks post-surgery, while promoting axonogenesis and remyelination at 8 weeks post-surgery, ultimately leading to enhanced motor functional recovery. This study elucidates the ability of neural regeneration strategy to promote motor functional recovery and provides a promising approach for designing multifunctional tissue for SCI treatment.
Collapse
Affiliation(s)
- Jiashang Liu
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomaterials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, PR China
| | - Ruijia Yan
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomaterials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, PR China
| | - Bixue Wang
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomaterials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, PR China
| | - Shu Chen
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomaterials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, PR China
| | - Hua Hong
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomaterials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, PR China
| | - Changsheng Liu
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomaterials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, PR China.
| | - Xi Chen
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomaterials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai 200237, PR China.
| |
Collapse
|
6
|
Picazo RA, Rojo C, Rodriguez-Quiros J, González-Gil A. Current Advances in Mesenchymal Stem Cell Therapies Applied to Wounds and Skin, Eye, and Neuromuscular Diseases in Companion Animals. Animals (Basel) 2024; 14:1363. [PMID: 38731367 PMCID: PMC11083242 DOI: 10.3390/ani14091363] [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: 04/10/2024] [Revised: 04/27/2024] [Accepted: 04/30/2024] [Indexed: 05/13/2024] Open
Abstract
Mesenchymal stem cells (MSCs) are considered a very promising alternative tool in cell therapies and regenerative medicine due to their ease of obtaining from various tissues and their ability to differentiate into different cell types. This manuscript provides a review of current knowledge on the use of MSC-based therapies as an alternative for certain common pathologies in dogs and cats where conventional treatments are ineffective. The aim of this review is to assist clinical veterinarians in making decisions about the suitability of each protocol from a clinical perspective, rather than focusing solely on research. MSC-based therapies have shown promising results in certain pathologies, such as spinal cord injuries, wounds, and skin and eye diseases. However, the effectiveness of these cell therapies can be influenced by a wide array of factors, leading to varying outcomes. Future research will focus on designing protocols and methodologies that allow more precise and effective MSC treatments for each case.
Collapse
Affiliation(s)
- Rosa Ana Picazo
- Department of Physiology, School of Veterinary Medicine, Complutense University of Madrid, 28040 Madrid, Spain;
| | - Concepción Rojo
- Department of Anatomy and Embryology, School of Veterinary Medicine, University Complutense of Madrid, 28040 Madrid, Spain;
| | - Jesus Rodriguez-Quiros
- Department of Animal Medicine and Surgery, School of Veterinary Medicine, Complutense University of Madrid, 28040 Madrid, Spain;
| | - Alfredo González-Gil
- Department of Physiology, School of Veterinary Medicine, Complutense University of Madrid, 28040 Madrid, Spain;
| |
Collapse
|
7
|
Jiu J, Liu H, Li D, Li J, Liu L, Yang W, Yan L, Li S, Zhang J, Li X, Li JJ, Wang B. 3D bioprinting approaches for spinal cord injury repair. Biofabrication 2024; 16:032003. [PMID: 38569491 DOI: 10.1088/1758-5090/ad3a13] [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: 10/10/2023] [Accepted: 04/03/2024] [Indexed: 04/05/2024]
Abstract
Regenerative healing of spinal cord injury (SCI) poses an ongoing medical challenge by causing persistent neurological impairment and a significant socioeconomic burden. The complexity of spinal cord tissue presents hurdles to successful regeneration following injury, due to the difficulty of forming a biomimetic structure that faithfully replicates native tissue using conventional tissue engineering scaffolds. 3D bioprinting is a rapidly evolving technology with unmatched potential to create 3D biological tissues with complicated and hierarchical structure and composition. With the addition of biological additives such as cells and biomolecules, 3D bioprinting can fabricate preclinical implants, tissue or organ-like constructs, andin vitromodels through precise control over the deposition of biomaterials and other building blocks. This review highlights the characteristics and advantages of 3D bioprinting for scaffold fabrication to enable SCI repair, including bottom-up manufacturing, mechanical customization, and spatial heterogeneity. This review also critically discusses the impact of various fabrication parameters on the efficacy of spinal cord repair using 3D bioprinted scaffolds, including the choice of printing method, scaffold shape, biomaterials, and biological supplements such as cells and growth factors. High-quality preclinical studies are required to accelerate the translation of 3D bioprinting into clinical practice for spinal cord repair. Meanwhile, other technological advances will continue to improve the regenerative capability of bioprinted scaffolds, such as the incorporation of nanoscale biological particles and the development of 4D printing.
Collapse
Affiliation(s)
- Jingwei Jiu
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
- Department of Orthopaedic Surgery, Shanxi Medical University Second Affiliated Hospital, Taiyuan, People's Republic of China
| | - Haifeng Liu
- Department of Orthopaedic Surgery, Shanxi Medical University Second Affiliated Hospital, Taiyuan, People's Republic of China
| | - Dijun Li
- Department of Orthopaedic Surgery, Shanxi Medical University Second Affiliated Hospital, Taiyuan, People's Republic of China
| | - Jiarong Li
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Lu Liu
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Wenjie Yang
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Lei Yan
- Department of Orthopaedic Surgery, Shanxi Medical University Second Affiliated Hospital, Taiyuan, People's Republic of China
| | - Songyan Li
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Jing Zhang
- Department of Emergency Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550001, People's Republic of China
| | - Xiaoke Li
- Department of Orthopaedic Surgery, Shanxi Medical University Second Affiliated Hospital, Taiyuan, People's Republic of China
| | - Jiao Jiao Li
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Bin Wang
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| |
Collapse
|
8
|
Iqbal J, Courville E, Kazim SF, Kogan M, Schmidt MH, Bowers CA. Role of nanotechnology in neurosurgery: A review of recent advances and their applications. World Neurosurg X 2024; 22:100298. [PMID: 38455250 PMCID: PMC10918265 DOI: 10.1016/j.wnsx.2024.100298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 02/20/2024] [Indexed: 03/09/2024] Open
Affiliation(s)
- Javed Iqbal
- School of Medicine, King Edward Medical University, Lahore, Pakistan
- Bowers Neurosurgical Frailty and Outcomes Data Science Lab, Albuquerque, NM, USA
| | - Evan Courville
- Department of Neurosurgery, University of New Mexico Hospital (UNMH), Albuquerque, NM, USA
- Bowers Neurosurgical Frailty and Outcomes Data Science Lab, Albuquerque, NM, USA
| | - Syed Faraz Kazim
- Department of Neurosurgery, University of New Mexico Hospital (UNMH), Albuquerque, NM, USA
- Bowers Neurosurgical Frailty and Outcomes Data Science Lab, Albuquerque, NM, USA
| | - Michael Kogan
- Bowers Neurosurgical Frailty and Outcomes Data Science Lab, Albuquerque, NM, USA
| | - Meic H. Schmidt
- Department of Neurosurgery, University of New Mexico Hospital (UNMH), Albuquerque, NM, USA
- Bowers Neurosurgical Frailty and Outcomes Data Science Lab, Albuquerque, NM, USA
| | - Christian A. Bowers
- Department of Neurosurgery, University of New Mexico Hospital (UNMH), Albuquerque, NM, USA
- Bowers Neurosurgical Frailty and Outcomes Data Science Lab, Albuquerque, NM, USA
| |
Collapse
|
9
|
Hersh AM, Weber-Levine C, Jiang K, Theodore N. Spinal Cord Injury: Emerging Technologies. Neurosurg Clin N Am 2024; 35:243-251. [PMID: 38423740 DOI: 10.1016/j.nec.2023.10.001] [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] [Indexed: 03/02/2024]
Abstract
The mainstay of treatment for spinal cord injury includes decompressive laminectomy and elevation of mean arterial pressure. However, outcomes often remain poor. Extensive research and ongoing clinical trials seek to design new treatment options for spinal cord injury, including stem cell therapy, scaffolds, brain-spine interfaces, exoskeletons, epidural electrical stimulation, ultrasound, and cerebrospinal fluid drainage. Some of these treatments are targeted at the initial acute window of injury, during which secondary damage occurs; others are designed to help patients living with chronic injuries.
Collapse
Affiliation(s)
- Andrew M Hersh
- Department of Neurosurgery, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Meyer 7-113, Baltimore, MD 21287, USA. https://twitter.com/AndrewMHersh
| | - Carly Weber-Levine
- Department of Neurosurgery, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Meyer 7-113, Baltimore, MD 21287, USA
| | - Kelly Jiang
- Department of Neurosurgery, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Meyer 7-113, Baltimore, MD 21287, USA. https://twitter.com/kellyjjiang
| | - Nicholas Theodore
- Department of Neurosurgery, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Meyer 7-113, Baltimore, MD 21287, USA; Orthopaedic Surgery & Biomedical Engineering, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
10
|
Chen Z, Sun Z, Fan Y, Yin M, Jin C, Guo B, Yin Y, Quan R, Zhao S, Han S, Cheng X, Liu W, Chen B, Xiao Z, Dai J, Zhao Y. Mimicked Spinal Cord Fibers Trigger Axonal Regeneration and Remyelination after Injury. ACS NANO 2023; 17:25591-25613. [PMID: 38078771 DOI: 10.1021/acsnano.3c09892] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2023]
Abstract
Spinal cord injury (SCI) causes tissue structure damage and composition changes of the neural parenchyma, resulting in severe consequences for spinal cord function. Mimicking the components and microstructure of spinal cord tissues holds promise for restoring the regenerative microenvironment after SCI. Here, we have utilized electrospinning technology to develop aligned decellularized spinal cord fibers (A-DSCF) without requiring synthetic polymers or organic solvents. A-DSCF preserves multiple types of spinal cord extracellular matrix proteins and forms a parallel-oriented structure. Compared to aligned collagen fibers (A-CF), A-DSCF exhibits stronger mechanical properties, improved enzymatic stability, and superior functionality in the adhesion, proliferation, axonal extension, and myelination of differentiated neural progenitor cells (NPCs). Notably, axon extension or myelination has been primarily linked to Agrin (AGRN), Laminin (LN), or Collagen type IV (COL IV) proteins in A-DSCF. When transplanted into rats with complete SCI, A-DSCF loaded with NPCs improves the survival, maturation, axon regeneration, and motor function of the SCI rats. These findings highlight the potential of structurally and compositionally biomimetic scaffolds to promote axonal extension and remyelination after SCI.
Collapse
Affiliation(s)
- Zhenni Chen
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zheng Sun
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yongheng Fan
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Man Yin
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chen Jin
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Bo Guo
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yanyun Yin
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Rui Quan
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuaijing Zhao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuyu Han
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaokang Cheng
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Weiyuan Liu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Bing Chen
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhifeng Xiao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jianwu Dai
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Yannan Zhao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
11
|
You Z, Gao X, Kang X, Yang W, Xiong T, Li Y, Wei F, Zhuang Y, Zhang T, Sun Y, Shen H, Dai J. Microvascular endothelial cells derived from spinal cord promote spinal cord injury repair. Bioact Mater 2023; 29:36-49. [PMID: 37621772 PMCID: PMC10444976 DOI: 10.1016/j.bioactmat.2023.06.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 06/06/2023] [Accepted: 06/23/2023] [Indexed: 08/26/2023] Open
Abstract
Neural regeneration after spinal cord injury (SCI) closely relates to the microvascular endothelial cell (MEC)-mediated neurovascular unit formation. However, the effects of central nerve system-derived MECs on neovascularization and neurogenesis, and potential signaling involved therein, are unclear. Here, we established a primary spinal cord-derived MECs (SCMECs) isolation with high cell yield and purity to describe the differences with brain-derived MECs (BMECs) and their therapeutic effects on SCI. Transcriptomics and proteomics revealed differentially expressed genes and proteins in SCMECs were involved in angiogenesis, immunity, metabolism, and cell adhesion molecular signaling was the only signaling pathway enriched of top 10 in differentially expressed genes and proteins KEGG analysis. SCMECs and BMECs could be induced angiogenesis by different stiffness stimulation of PEG hydrogels with elastic modulus 50-1650 Pa for SCMECs and 50-300 Pa for BMECs, respectively. Moreover, SCMECs and BMECs promoted spinal cord or brain-derived NSC (SNSC/BNSC) proliferation, migration, and differentiation at different levels. At certain dose, SCMECs in combination with the NeuroRegen scaffold, showed higher effectiveness in the promotion of vascular reconstruction. The potential underlying mechanism of this phenomenon may through VEGF/AKT/eNOS- signaling pathway, and consequently accelerated neuronal regeneration and functional recovery of SCI rats compared to BMECs. Our findings suggested a promising role of SCMECs in restoring vascularization and neural regeneration.
Collapse
Affiliation(s)
- Zhifeng You
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Xu Gao
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
- Department of Orthopaedic Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - Xinyi Kang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Wen Yang
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Tiandi Xiong
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, China
| | - Yue Li
- i-Lab, Key Laboratory of Multifunction Nanomaterials and Smart Systems, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Feng Wei
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, China
| | - Yan Zhuang
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, China
| | - Ting Zhang
- i-Lab, Key Laboratory of Multifunction Nanomaterials and Smart Systems, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Yifu Sun
- Department of Orthopaedic Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - He Shen
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, China
| | - Jianwu Dai
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| |
Collapse
|
12
|
Jiang W, Zhang X, Yu S, Yan F, Chen J, Liu J, Dong C. Decellularized extracellular matrix in the treatment of spinal cord injury. Exp Neurol 2023; 368:114506. [PMID: 37597763 DOI: 10.1016/j.expneurol.2023.114506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/04/2023] [Accepted: 08/15/2023] [Indexed: 08/21/2023]
Abstract
Functional limitation caused by spinal cord injury (SCI) has the problem of significant clinical and economic burden. Damaged spinal axonal connections and an inhibitory environment severely hamper neuronal function. Regenerative biomaterials can fill the cavity and produce an optimal microenvironment at the site of SCI, inhibiting apoptosis, inflammation, and glial scar formation while promoting neurogenesis, axonal development, and angiogenesis. Decellularization aims to eliminate cells from the ultrastructure of tissues while keeping tissue-specific components that are similar to the structure of real tissues, making decellularized extracellular matrix (dECM) a suitable scaffold for tissue engineering. dECM has good biocompatibility, it can be widely obtained from natural organs of different species, and can be co-cultured with cells for 3D printing to obtain the target scaffold. In this paper, we reviewed the pathophysiology of SCI, the characteristics of dECM and its preparation method, and the application of dECM in the treatment of SCI. Although dECM has shown its therapeutic effect at present, there are still many indicators that need to be taken into account, such as the difficulty in obtaining materials and standardized production mode for large-scale use, the effect of decellularization on the physical and chemical properties of dECM, and the study on the synergistic effect of dECM and cells.
Collapse
Affiliation(s)
- Wenwei Jiang
- Department of Anatomy, Medical College of Nantong University, Nantong, China
| | - Xuanxuan Zhang
- Department of Anatomy, Medical College of Nantong University, Nantong, China
| | - Shumin Yu
- Department of Anatomy, Medical College of Nantong University, Nantong, China
| | - Fangsu Yan
- Department of Anatomy, Medical College of Nantong University, Nantong, China
| | - Jiaxi Chen
- Department of Anatomy, Medical College of Nantong University, Nantong, China
| | - Jinyi Liu
- Department of Anatomy, Medical College of Nantong University, Nantong, China
| | - Chuanming Dong
- Department of Anatomy, Medical College of Nantong University, Nantong, China; Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, Jiangsu Province, China.
| |
Collapse
|
13
|
Xu B, Liu D, Liu W, Long G, Liu W, Wu Y, He X, Shen Y, Jiang P, Yin M, Fan Y, Shen H, Shi L, Zhang Q, Xue W, Jin C, Chen Z, Chen B, Li J, Hu Y, Li X, Xiao Z, Zhao Y, Dai J. Engineered human spinal cord-like tissues with dorsal and ventral neuronal progenitors for spinal cord injury repair in rats and monkeys. Bioact Mater 2023; 27:125-137. [PMID: 37064803 PMCID: PMC10090126 DOI: 10.1016/j.bioactmat.2023.03.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 03/05/2023] [Accepted: 03/21/2023] [Indexed: 03/31/2023] Open
Abstract
Transplanting human neural progenitor cells is a promising method of replenishing the lost neurons after spinal cord injury (SCI), but differentiating neural progenitor cells into the diverse types of mature functional spinal cord neurons in vivo is challenging. In this study, engineered human embryonic spinal cord-like tissues with dorsal and ventral neuronal characters (DV-SC) were generated by inducing human neural progenitor cells (hscNPCs) to differentiate into various types of dorsal and ventral neuronal cells on collagen scaffold in vitro. Transplantation of DV-SC into complete SCI models in rats and monkeys showed better therapeutic effects than undifferentiated hscNPCs, including pronounced cell survival and maturation. DV-SC formed a targeted connection with the host's ascending and descending axons, partially restored interrupted neural circuits, and improved motor evoked potentials and the hindlimb function of animals with SCI. This suggests that the transplantation of pre-differentiated hscNPCs with spinal cord dorsal and ventral neuronal characteristics could be a promising strategy for SCI repair.
Collapse
|
14
|
Feng F, Song X, Tan Z, Tu Y, Xiao L, Xie P, Ma Y, Sun X, Ma J, Rong L, He L. Cooperative assembly of a designer peptide and silk fibroin into hybrid nanofiber gels for neural regeneration after spinal cord injury. SCIENCE ADVANCES 2023; 9:eadg0234. [PMID: 37352345 PMCID: PMC10289662 DOI: 10.1126/sciadv.adg0234] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 05/17/2023] [Indexed: 06/25/2023]
Abstract
Local reconstruction of a permissive environment with biomaterials is a promising strategy to treat spinal cord injury (SCI). We reported a hybrid hydrogel fabricated from a small functional self-assembling peptide (F-SAP) and large silk fibroin (SF). The diffusion of SF micelles into F-SAP solution was driven by the dynamic synergy between osmotic pressure and F-SAP/SF electrostatic interactions, resulting in the rearrangement of SF micelles and the formation of rod-like filaments with axes nearly perpendicular to F-SAP nanofibers. Spectroscopy analysis, including circular dichroism, Raman and fluorescence, indicated conformation changes of SF from random coil to β sheet, which contributed to enhanced mechanical properties of the resultant hybrid hydrogel. Furthermore, the F-SAP/SF hybrid hydrogel coupled with controlled release of NT-3 provided a permissive environment for neural regeneration by providing nanofibrous substrates for regenerating axons, inflammatory modulation and remyelination, consequently resulting in improved locomotion and electrophysiological properties. This hydrogel could be used as a long-term stent in vivo for the treatment of SCI.
Collapse
Affiliation(s)
- Feng Feng
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Xiyong Song
- Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki 00014, Finland
| | - Zan Tan
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Yujie Tu
- College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Longyou Xiao
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Pengfei Xie
- College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Yahao Ma
- College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Xiumin Sun
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Junwu Ma
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Limin Rong
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Liumin He
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| |
Collapse
|
15
|
BMP-2 functional polypeptides relieve osteolysis via bi-regulating bone formation and resorption coupled with macrophage polarization. NPJ Regen Med 2023; 8:6. [PMID: 36759627 PMCID: PMC9911742 DOI: 10.1038/s41536-023-00279-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 01/25/2023] [Indexed: 02/11/2023] Open
Abstract
Osteolysis caused by wear debris around the prosthesis is the main reason for aseptic loosening. Extending prosthetic service life is still challenging. In this study, we first synthesized a bone morphogenetic protein-2 (BMP-2) functional polypeptide (BMP2pp), and evaluated the effects of BMP2pp on macrophage polarization and impaired osteogenesis caused by titanium (Ti) particles in vitro. Then, we delineated the impact of BMP2pp on bone formation and resorption in a mouse calvarial bone osteolysis model induced by Ti particles. The results showed that BMP2pp not only alleviated the Ti-induced inhibition of osteoblastic differentiation in human placenta-derived mesenchymal stem cells (hPMSCs) but also prevented Ti-induced M1 macrophage polarization and promoted M2 macrophage differentiation in mice. Conditioned medium from BMP2pp-activated macrophages increased the osteogenesis of hPMSCs. The western blot results indicated a significant decrease in the expression of NF-κB inducing kinase (NIK) and phospho-NF-κB p65 in bone marrow-derived macrophages treated with BMP2pp. Furthermore, we clarified the protective effect of BMP2pp on bone formation and the reduction in bone resorption coupled with the immunomodulatory properties of calvarial osteolysis in mice. In summary, BMP2pp ameliorated the Ti-mediated impairment in osteogenic potential of hPMSCs, suppressed the M1 polarization of macrophages by inhibiting the activation of the NF-κB signaling pathway, and ameliorated Ti-induced bone osteolysis. Our research suggests that BMP2pp may be a potential option for treating prosthetic loosening induced by wear debris from prostheses.
Collapse
|
16
|
Feng C, Deng L, Yong YY, Wu JM, Qin DL, Yu L, Zhou XG, Wu AG. The Application of Biomaterials in Spinal Cord Injury. Int J Mol Sci 2023; 24:816. [PMID: 36614259 PMCID: PMC9821025 DOI: 10.3390/ijms24010816] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/19/2022] [Accepted: 12/30/2022] [Indexed: 01/05/2023] Open
Abstract
The spinal cord and the brain form the central nervous system (CNS), which is the most important part of the body. However, spinal cord injury (SCI) caused by external forces is one of the most difficult types of neurological injury to treat, resulting in reduced or even absent motor, sensory and autonomic functions. It leads to the reduction or even disappearance of motor, sensory and self-organizing nerve functions. Currently, its incidence is increasing each year worldwide. Therefore, the development of treatments for SCI is urgently needed in the clinic. To date, surgery, drug therapy, stem cell transplantation, regenerative medicine, and rehabilitation therapy have been developed for the treatment of SCI. Among them, regenerative biomaterials that use tissue engineering and bioscaffolds to transport cells or drugs to the injured site are considered the most promising option. In this review, we briefly introduce SCI and its molecular mechanism and summarize the application of biomaterials in the repair and regeneration of tissue in various models of SCI. However, there is still limited evidence about the treatment of SCI with biomaterials in the clinic. Finally, this review will provide inspiration and direction for the future study and application of biomaterials in the treatment of SCI.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xiao-Gang Zhou
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - An-Guo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
17
|
Gao X, You Z, Li Y, Kang X, Yang W, Wang H, Zhang T, Zhao X, Sun Y, Shen H, Dai J. Multifunctional hydrogel modulates the immune microenvironment to improve allogeneic spinal cord tissue survival for complete spinal cord injury repair. Acta Biomater 2023; 155:235-246. [PMID: 36384221 DOI: 10.1016/j.actbio.2022.11.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 10/21/2022] [Accepted: 11/08/2022] [Indexed: 11/14/2022]
Abstract
Transplantation of allogeneic adult spinal cord tissues (aSCTs) to replace the injured spinal cord, serves as a promising strategy in complete spinal cord injury (SCI) repair. However, in addition to allograft immune rejection, damage-associated molecular pattern (DAMP)-mediated inflammatory microenvironments greatly impair the survival and function of transplants. In this study, we aimed to regulate the immune microenvironment after aSCT implantation by developing a functional hybrid gelatin and hyaluronic acid hydrogel (F-G/H) modified with cationic polymers and anti-inflammatory cytokines that can gelatinize at both ends of the aSCT to glue the grafts for perfect matching at defects. The F-G/H hydrogel exhibited the capacities of DAMP scavenging, sustainably released anti-inflammatory cytokines, and reduced lymphocyte accumulation, thereby modulating the immune response and enhancing the survival and function of aSCTs. When the hydrogel was used in combination with a systemic immunosuppressive drug treatment, the locomotor functions of SCI rats were significantly improved after aSCTs and F-G/H transplantation. This biomaterial-based immunomodulatory strategy may provide the potential for spinal cord graft replacement for treating SCI. STATEMENT OF SIGNIFICANCE: In this study, we aimed to regulate the immune microenvironment by developing a functional hybrid gelatin and hyaluronic acid hydrogel (F-G/H) modified with cationic polymers and anti-inflammatory cytokines that can gelatinize at both ends of the aSCT to glue the grafts for perfect matching at defects. We found that with the treatment of F-G/H hydrogel, the aSCT survival and function was significantly improved, as a result of reducing recruitment and activation of immune cells through TLR- and ST-2- related signaling. With the combination of immunosuppressive drug treatment, the locomotor functions of SCI rats were significantly improved after aSCTs and F-G/H transplantation. Findings from this work suggest the potential application of the F-G/H as a biomaterial-based immunoregulatory strategy for improving the therapeutic efficiency of the transplanted spinal cord graft for spinal cord injury repair.
Collapse
Affiliation(s)
- Xu Gao
- Department of Orthopaedic Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, P. R. China; Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of NanoTech and NanoBionics, Chinese Academy of Sciences, Suzhou, 215123, P. R. China
| | - Zhifeng You
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of NanoTech and NanoBionics, Chinese Academy of Sciences, Suzhou, 215123, P. R. China
| | - Yue Li
- i-Lab, Key Laboratory of Multifunction Nanomaterials and Smart Systems, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, P. R. China
| | - Xinyi Kang
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of NanoTech and NanoBionics, Chinese Academy of Sciences, Suzhou, 215123, P. R. China
| | - Wen Yang
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of NanoTech and NanoBionics, Chinese Academy of Sciences, Suzhou, 215123, P. R. China
| | - Huiru Wang
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of NanoTech and NanoBionics, Chinese Academy of Sciences, Suzhou, 215123, P. R. China; School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China
| | - Ting Zhang
- i-Lab, Key Laboratory of Multifunction Nanomaterials and Smart Systems, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, P. R. China
| | - Xinhao Zhao
- Department of Orthopaedic Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, P. R. China; State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, P. R. China
| | - Yifu Sun
- Department of Orthopaedic Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, P. R. China.
| | - He Shen
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of NanoTech and NanoBionics, Chinese Academy of Sciences, Suzhou, 215123, P. R. China; School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China.
| | - Jianwu Dai
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of NanoTech and NanoBionics, Chinese Academy of Sciences, Suzhou, 215123, P. R. China; School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, China; State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, P. R. China.
| |
Collapse
|
18
|
Blando S, Anchesi I, Mazzon E, Gugliandolo A. Can a Scaffold Enriched with Mesenchymal Stem Cells Be a Good Treatment for Spinal Cord Injury? Int J Mol Sci 2022; 23:ijms23147545. [PMID: 35886890 PMCID: PMC9319719 DOI: 10.3390/ijms23147545] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 07/01/2022] [Accepted: 07/05/2022] [Indexed: 12/10/2022] Open
Abstract
Spinal cord injury (SCI) is a worldwide highly crippling disease that can lead to the loss of motor and sensory neurons. Among the most promising therapies, there are new techniques of tissue engineering based on stem cells that promote neuronal regeneration. Among the different types of stem cells, mesenchymal stem cells (MSCs) seem the most promising. Indeed, MSCs are able to release trophic factors and to differentiate into the cell types that can be found in the spinal cord. Currently, the most common procedure to insert cells in the lesion site is infusion. However, this causes a low rate of survival and engraftment in the lesion site. For these reasons, tissue engineering is focusing on bioresorbable scaffolds to help the cells to stay in situ. Scaffolds do not only have a passive role but become fundamental for the trophic support of cells and the promotion of neuroregeneration. More and more types of materials are being studied as scaffolds to decrease inflammation and increase the engraftment as well as the survival of the cells. Our review aims to highlight how the use of scaffolds made from biomaterials enriched with MSCs gives positive results in in vivo SCI models as well as the first evidence obtained in clinical trials.
Collapse
|
19
|
Fan C, Yang W, Zhang L, Cai H, Zhuang Y, Chen Y, Zhao Y, Dai J. Restoration of spinal cord biophysical microenvironment for enhancing tissue repair by injury-responsive smart hydrogel. Biomaterials 2022; 288:121689. [DOI: 10.1016/j.biomaterials.2022.121689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 06/10/2022] [Accepted: 07/18/2022] [Indexed: 11/02/2022]
|
20
|
Zachariou D, Evangelopoulos DS, Rozis M, Papagrigorakis E, Galanis A, Vavourakis M, Pneumaticos SG, Vlamis J. Application of Collagen-Based Scaffolds for the Treatment of Spinal Cord Injuries in Animal Models: A Literature Update. Cureus 2022; 14:e25997. [PMID: 35859951 PMCID: PMC9288156 DOI: 10.7759/cureus.25997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2022] [Indexed: 11/05/2022] Open
Abstract
SCI is regarded as one of the most devastating central nervous system (CNS) injuries, exhibiting an alarmingly rising incidence rate, indirectly connected with the expansion of the global economy. The consequences of SCI are multidimensional: SCI injuries may result in permanent voluntary motor dysfunction and loss of sensation while incurring heavy economic and psychological burdens as part of the treatment. Thus, it is crucial to develop effective and suitable SCI treatment strategies. Collagen-based scaffold application is one of the most promising methods of SCI treatment. This review compiles newer bibliographical data regarding the application of collagen scaffolds for the treatment of Spinal cord injury (SCI) in animal models. Recently, several relevant studies have been carried out using carefully selected animals with similar pathophysiology to humans. In mouse, rat and canine models that have undergone transection or hemisection, the stump connection, the transplanted cell differentiation, and the elimination of glial scar are promising. Also, encouraging results have been found regarding the increased neuronal growth, the decreased collagen deposition, the behavioral recovery, the improved electrophysiology, and the enhanced axonal regeneration.
Collapse
|
21
|
Wang H, Xia Y, Li B, Li Y, Fu C. Reverse Adverse Immune Microenvironments by Biomaterials Enhance the Repair of Spinal Cord Injury. Front Bioeng Biotechnol 2022; 10:812340. [PMID: 35646849 PMCID: PMC9136098 DOI: 10.3389/fbioe.2022.812340] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 04/29/2022] [Indexed: 12/14/2022] Open
Abstract
Spinal cord injury (SCI) is a severe and traumatic disorder that ultimately results in the loss of motor, sensory, and autonomic nervous function. After SCI, local immune inflammatory response persists and does not weaken or disappear. The interference of local adverse immune factors after SCI brings great challenges to the repair of SCI. Among them, microglia, macrophages, neutrophils, lymphocytes, astrocytes, and the release of various cytokines, as well as the destruction of the extracellular matrix are mainly involved in the imbalance of the immune microenvironment. Studies have shown that immune remodeling after SCI significantly affects the survival and differentiation of stem cells after transplantation and the prognosis of SCI. Recently, immunological reconstruction strategies based on biomaterials have been widely explored and achieved good results. In this review, we discuss the important factors leading to immune dysfunction after SCI, such as immune cells, cytokines, and the destruction of the extracellular matrix. Additionally, the immunomodulatory strategies based on biomaterials are summarized, and the clinical application prospects of these immune reconstructs are evaluated.
Collapse
|
22
|
Kaminska A, Radoszkiewicz K, Rybkowska P, Wedzinska A, Sarnowska A. Interaction of Neural Stem Cells (NSCs) and Mesenchymal Stem Cells (MSCs) as a Promising Approach in Brain Study and Nerve Regeneration. Cells 2022; 11:cells11091464. [PMID: 35563770 PMCID: PMC9105617 DOI: 10.3390/cells11091464] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/20/2022] [Accepted: 04/22/2022] [Indexed: 11/16/2022] Open
Abstract
Rapid developments in stem cell research in recent years have provided a solid foundation for their use in medicine. Over the last few years, hundreds of clinical trials have been initiated in a wide panel of indications. Disorders and injuries of the nervous system still remain a challenge for the regenerative medicine. Neural stem cells (NSCs) are the optimal cells for the central nervous system restoration as they can differentiate into mature cells and, most importantly, functional neurons and glial cells. However, their application is limited by multiple factors such as difficult access to source material, limited cells number, problematic, long and expensive cultivation in vitro, and ethical considerations. On the other hand, according to the available clinical databases, most of the registered clinical trials involving cell therapies were carried out with the use of mesenchymal stem/stromal/signalling cells (MSCs) obtained from afterbirth or adult human somatic tissues. MSCs are the multipotent cells which can also differentiate into neuron-like and glia-like cells under proper conditions in vitro; however, their main therapeutic effect is more associated with secretory and supportive properties. MSCs, as a natural component of cell niche, affect the environment through immunomodulation as well as through the secretion of the trophic factors. In this review, we discuss various therapeutic strategies and activated mechanisms related to bilateral MSC–NSC interactions, differentiation of MSCs towards the neural cells (subpopulation of crest-derived cells) under the environmental conditions, bioscaffolds, or co-culture with NSCs by recreating the conditions of the neural cell niche.
Collapse
|
23
|
|
24
|
Van den Bos J, Ouaamari YE, Wouters K, Cools N, Wens I. Are Cell-Based Therapies Safe and Effective in the Treatment of Neurodegenerative Diseases? A Systematic Review with Meta-Analysis. Biomolecules 2022; 12:340. [PMID: 35204840 PMCID: PMC8869169 DOI: 10.3390/biom12020340] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/14/2022] [Accepted: 02/17/2022] [Indexed: 12/13/2022] Open
Abstract
Over the past two decades, significant advances have been made in the field of regenerative medicine. However, despite being of the utmost clinical urgency, there remains a paucity of therapeutic strategies for conditions with substantial neurodegeneration such as (progressive) multiple sclerosis (MS), spinal cord injury (SCI), Parkinson's disease (PD) and Alzheimer's disease (AD). Different cell types, such as mesenchymal stromal cells (MSC), neuronal stem cells (NSC), olfactory ensheathing cells (OEC), neurons and a variety of others, already demonstrated safety and regenerative or neuroprotective properties in the central nervous system during the preclinical phase. As a result of these promising findings, in recent years, these necessary types of cell therapies have been intensively tested in clinical trials to establish whether these results could be confirmed in patients. However, extensive research is still needed regarding elucidating the exact mechanism of action, possible immune rejection, functionality and survival of the administered cells, dose, frequency and administration route. To summarize the current state of knowledge, we conducted a systematic review with meta-analysis. A total of 27,043 records were reviewed by two independent assessors and 71 records were included in the final quantitative analysis. These results show that the overall frequency of serious adverse events was low: 0.03 (95% CI: 0.01-0.08). In addition, several trials in MS and SCI reported efficacy data, demonstrating some promising results on clinical outcomes. All randomized controlled studies were at a low risk of bias due to appropriate blinding of the treatment, including assessors and patients. In conclusion, cell-based therapies in neurodegenerative disease are safe and feasible while showing promising clinical improvements. Nevertheless, given their high heterogeneity, the results require a cautious approach. We advocate for the harmonization of study protocols of trials investigating cell-based therapies in neurodegenerative diseases, adverse event reporting and investigation of clinical outcomes.
Collapse
Affiliation(s)
- Jasper Van den Bos
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Universiteitsplein 1, B-2610 Antwerpen, Belgium; (Y.E.O.); (N.C.); (I.W.)
| | - Yousra El Ouaamari
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Universiteitsplein 1, B-2610 Antwerpen, Belgium; (Y.E.O.); (N.C.); (I.W.)
| | - Kristien Wouters
- Clinical Trial Center (CTC), CRC Antwerp, Antwerp University Hospital, University of Antwerp, Drie Eikenstraat 655, B-2650 Edegem, Belgium;
| | - Nathalie Cools
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Universiteitsplein 1, B-2610 Antwerpen, Belgium; (Y.E.O.); (N.C.); (I.W.)
- Center for Cell Therapy and Regenerative Medicine (CCRG), Antwerp University Hospital, Drie Eikenstraat 655, B-2650 Edegem, Belgium
| | - Inez Wens
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Universiteitsplein 1, B-2610 Antwerpen, Belgium; (Y.E.O.); (N.C.); (I.W.)
| |
Collapse
|
25
|
Deng WS, Liu XY, Ma K, Liang B, Liu YF, Wang RJ, Chen XY, Zhang S. Recovery of motor function in rats with complete spinal cord injury following implantation of collagen/silk fibroin scaffold combined with human umbilical cord-mesenchymal stem cells. Rev Assoc Med Bras (1992) 2021; 67:1342-1348. [PMID: 34816932 DOI: 10.1590/1806-9282.20200697] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 10/08/2020] [Indexed: 02/08/2023] Open
Abstract
OBJECTIVE This study aimed to assess the effect of the collagen/silk fibroin scaffolds seeded with human umbilical cord-mesenchymal stem cells on functional recovery after acute complete spinal cord injury. METHODS The fibroin and collagen were mixed (mass ratio, 3:7), and the composite scaffolds were produced. Forty rats were randomly divided into the Sham group (without spinal cord injury), spinal cord injury group (spinal cord transection without any implantation), collagen/silk fibroin scaffolds group (spinal cord transection with implantation of the collagen/silk fibroin scaffolds), and collagen/silk fibroin scaffolds + human umbilical cord-mesenchymal stem cells group (spinal cord transection with the implantation of the collagen/silk fibroin scaffolds co-cultured with human umbilical cord-mesenchymal stem cells). Motor evoked potential, Basso-Beattie-Bresnahan scale, modified Bielschowsky's silver staining, and immunofluorescence staining were performed. RESULTS The BBB scores in the collagen/silk fibroin scaffolds + human umbilical cord-mesenchymal stem cells group were significantly higher than those in the spinal cord injury and collagen/silk fibroin scaffolds groups (p<0.05 or p<0.01). The amplitude and latency were markedly improved in the collagen/silk fibroin scaffolds + human umbilical cord-mesenchymal stem cells group compared with the spinal cord injury and collagen/silk fibroin scaffolds groups (p<0.05 or p<0.01). Meanwhile, compared to the spinal cord injury and collagen/silk fibroin scaffolds groups, more neurofilament positive nerve fiber ensheathed by myelin basic protein positive structure at the injury site were observed in the collagen/silk fibroin scaffolds + human umbilical cord-mesenchymal stem cells group (p<0.01, p<0.05). The results of Bielschowsky's silver staining indicated more nerve fibers was observed at the lesion site in the collagen/silk fibroin scaffolds + human umbilical cord-mesenchymal stem cells group compared with the spinal cord injury and collagen/silk fibroin scaffolds groups (p<0.01, p< 0.05). CONCLUSION The results demonstrated that the transplantation of human umbilical cord-mesenchymal stem cells on a collagen/silk fibroin scaffolds could promote nerve regeneration, and recovery of neurological function after acute spinal cord injury.
Collapse
Affiliation(s)
- Wu-Sheng Deng
- Gansu University of Chinese Medicine, College of Integrated Traditional Chinese and Western Medicine - Gansu Province, China
| | - Xiao-Yin Liu
- Pingjin Hospital Brain Center, Tianjin Key Laboratory of Neurotrauma Repair, Characteristic Medical Center of Chinese People's Armed Police Force - Tianjin, China.,Sichuan University, West China Hospital, Department of Neurosurgery - Chengdu, China
| | - Ke Ma
- Pingjin Hospital Brain Center, Tianjin Key Laboratory of Neurotrauma Repair, Characteristic Medical Center of Chinese People's Armed Police Force - Tianjin, China
| | - Bing Liang
- Pingjin Hospital Brain Center, Tianjin Key Laboratory of Neurotrauma Repair, Characteristic Medical Center of Chinese People's Armed Police Force - Tianjin, China
| | - Ying-Fu Liu
- Cangzhou nanobody technology innovation center - Cangzhou, China
| | - Ren-Jie Wang
- Pingjin Hospital Brain Center, Tianjin Key Laboratory of Neurotrauma Repair, Characteristic Medical Center of Chinese People's Armed Police Force - Tianjin, China
| | - Xu-Yi Chen
- Pingjin Hospital Brain Center, Tianjin Key Laboratory of Neurotrauma Repair, Characteristic Medical Center of Chinese People's Armed Police Force - Tianjin, China
| | - Sai Zhang
- Pingjin Hospital Brain Center, Tianjin Key Laboratory of Neurotrauma Repair, Characteristic Medical Center of Chinese People's Armed Police Force - Tianjin, China
| |
Collapse
|
26
|
The Unique Properties of Placental Mesenchymal Stromal Cells: A Novel Source of Therapy for Congenital and Acquired Spinal Cord Injury. Cells 2021; 10:cells10112837. [PMID: 34831060 PMCID: PMC8616037 DOI: 10.3390/cells10112837] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/15/2021] [Accepted: 10/19/2021] [Indexed: 12/28/2022] Open
Abstract
Spinal cord injury (SCI) is a devasting condition with no reliable treatment. Spina bifida is the most common cause of congenital SCI. Cell-based therapies using mesenchymal stem/stromal cells (MSCS) have been largely utilized in SCI. Several clinical trials for acquired SCI use adult tissue-derived MSC sources, including bone-marrow, adipose, and umbilical cord tissues. The first stem/stromal cell clinical trial for spina bifida is currently underway (NCT04652908). The trial uses early gestational placental-derived mesenchymal stem/stromal cells (PMSCs) during the fetal repair of myelomeningocele. PMSCs have been shown to exhibit unique neuroprotective, angiogenic, and antioxidant properties, all which are promising applications for SCI. This review will summarize the unique properties and current applications of PMSCs and discuss their therapeutic role for acquired SCI.
Collapse
|
27
|
Chen X, Wang Y, Zhou G, Hu X, Han S, Gao J. The combination of nanoscaffolds and stem cell transplantation: Paving a promising road for spinal cord injury regeneration. Biomed Pharmacother 2021; 143:112233. [PMID: 34649357 DOI: 10.1016/j.biopha.2021.112233] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/05/2021] [Accepted: 09/19/2021] [Indexed: 12/14/2022] Open
Abstract
Spinal cord injury (SCI), one of the most devastating traumas, has caused long-term disability in millions of people worldwide. The pathophysiology of SCI primarily occurs in two stages classified as primary injury and secondary injury. Due to the rupture of axons and the apoptosis of neurons, patients lose their motor, sensory, and reflex functions, which also imposes a huge burden on families and society. However, traditional surgery does not facilitate neuronal regeneration. Although neural stem cells (NSCs) have the potential for multidirectional differentiation, the probability of differentiation into neurons and survival are still low. Surprisingly, the unique properties of nanotechnologies enable targeted drug delivery while reducing adverse reactions, assisting NSCs in differentiating into neurons. Here, recent studies on promising nanoscaffolds are highlighted, and their strengths and drawbacks are evaluated. Although the treatment of SCI remains fraught with challenges, the combination of nanoscaffolds and NSCs pave a promising road for SCI regeneration.
Collapse
Affiliation(s)
- Xiaokun Chen
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yiyang Wang
- School of Medicine, Tsinghua University, Haidian District, Beijing, China
| | - Gang Zhou
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xianghui Hu
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shiyuan Han
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jun Gao
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
28
|
Deng WS, Yang K, Liang B, Liu YF, Chen XY, Zhang S. Collagen/heparin sulfate scaffold combined with mesenchymal stem cells treatment for canines with spinal cord injury: A pilot feasibility study. J Orthop Surg (Hong Kong) 2021; 29:23094990211012293. [PMID: 34060363 DOI: 10.1177/23094990211012293] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Due to endogenous neuronal deficiency and glial scar formation, spinal cord injury (SCI) often leads to irreversible neurological loss. Accumulating evidence has shown that a suitable scaffold has important value for promoting nerve regeneration after SCI. Collagen/heparin sulfate scaffold (CHSS) has shown effect for guiding axonal regeneration and decreasing glial scar deposition after SCI. The current research aimed to evaluate the utility of the CHSSs adsorbed with mesenchymal stem cells (MSCs) on nerve regeneration, and functional recovery after acute complete SCI. METHODS CHSSs were prepared, and evaluated for biocompatibility. The CHSSs adsorbed with MSCs were transplanted into these canines with complete SCI. RESULTS We observed that MSCs had good biocompatibility with CHSSs. In complete transverse SCI models, the implantation of CHSS co-cultured with MSCs exhibited significant improvement in locomotion, motor evoked potential, magnetic resonance imaging, diffusion tensor imaging, and urodynamic parameters. Meanwhile, nerve fibers were markedly improved in the CHSS adsorbed with MSCs group. Moreover, we observed that the implantation of CHSS combined with MSCs modulated inflammatory cytokine levels. CONCLUSIONS The results preliminarily demonstrated that the transplantation of MSCs on a CHSS could improve the recovery of motor function after SCI. Thus, implanting the MSCs-laden CHSS is a promising combinatorial therapy for treatment in acute SCI.
Collapse
Affiliation(s)
- Wu-Sheng Deng
- College of Integrated Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu Province, China
| | - Kun Yang
- The First Affiliated Hospital of Hainan Medical College, Haikou, China
| | - Bing Liang
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of Chinese People's Armed Police Force, Tianjin, China
| | - Ying-Fu Liu
- Cang Zhou Nanobody Technology Innovation Center, Cangzhou, China
| | - Xu-Yi Chen
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of Chinese People's Armed Police Force, Tianjin, China
| | - Sai Zhang
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of Chinese People's Armed Police Force, Tianjin, China
| |
Collapse
|
29
|
Tang F, Tang J, Zhao Y, Zhang J, Xiao Z, Chen B, Han G, Yin N, Jiang X, Zhao C, Cheng S, Wang Z, Chen Y, Chen Q, Song K, Zhang Z, Niu J, Wang L, Shi Q, Chen L, Yang H, Hou S, Zhang S, Dai J. Long-term clinical observation of patients with acute and chronic complete spinal cord injury after transplantation of NeuroRegen scaffold. SCIENCE CHINA-LIFE SCIENCES 2021; 65:909-926. [PMID: 34406569 DOI: 10.1007/s11427-021-1985-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 06/08/2021] [Indexed: 02/05/2023]
Abstract
Spinal cord injury (SCI) often results in an inhibitory environment at the injury site. In our previous studies, transplantation of a scaffold combined with stem cells was proven to induce neural regeneration in animal models of complete SCI. Based on these preclinical studies, collagen scaffolds loaded with the patients' own bone marrow mononuclear cells or human umbilical cord mesenchymal stem cells were transplanted into SCI patients. Fifteen patients with acute complete SCI and 51 patients with chronic complete SCI were enrolled and followed up for 2 to 5 years. No serious adverse events related to functional scaffold transplantation were observed. Among the patients with acute SCI, five patients achieved expansion of their sensory positions and six patients recovered sensation in the bowel or bladder. Additionally, four patients regained voluntary walking ability accompanied by reconnection of neural signal transduction. Among patients with chronic SCI, 16 patients achieved expansion of their sensation level and 30 patients experienced enhanced reflexive defecation sensation or increased skin sweating below the injury site. Nearly half of the patients with chronic cervical SCI developed enhanced finger activity. These long-term follow-up results suggest that functional scaffold transplantation may represent a feasible treatment for patients with complete SCI.
Collapse
Affiliation(s)
- Fengwu Tang
- Characteristics Medical Center of the Chinese People's Armed Police Forces (CAPF), Tianjin, 300162, China
| | - Jiaguang Tang
- Fourth Medical Center of People's Liberation Army (PLA) General Hospital, Beijing, 100048, China.,Department of Orthopaedics, Beijing Tongren Hospital, Beijing, 100730, China
| | - Yannan Zhao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100190, China.,Beijing ZhongKeZaiKang Biotechnology Co., Ltd, Beijing, 101407, China
| | - Jiaojiao Zhang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100190, China
| | - Zhifeng Xiao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100190, China.,Beijing ZhongKeZaiKang Biotechnology Co., Ltd, Beijing, 101407, China
| | - Bing Chen
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100190, China
| | - Guang Han
- Characteristics Medical Center of the Chinese People's Armed Police Forces (CAPF), Tianjin, 300162, China
| | - Na Yin
- Characteristics Medical Center of the Chinese People's Armed Police Forces (CAPF), Tianjin, 300162, China.,Department of Rehabilitation, the 983rd Hospital of the Joint Logistics Support Force of the Chinese People's Liberation Army, Tianjin, 300141, China
| | - Xianfeng Jiang
- Characteristics Medical Center of the Chinese People's Armed Police Forces (CAPF), Tianjin, 300162, China
| | - Changyu Zhao
- Characteristics Medical Center of the Chinese People's Armed Police Forces (CAPF), Tianjin, 300162, China
| | - Shixiang Cheng
- Characteristics Medical Center of the Chinese People's Armed Police Forces (CAPF), Tianjin, 300162, China
| | - Ziqiang Wang
- Fourth Medical Center of People's Liberation Army (PLA) General Hospital, Beijing, 100048, China
| | - Yumei Chen
- Fourth Medical Center of People's Liberation Army (PLA) General Hospital, Beijing, 100048, China
| | - Qiaoling Chen
- Fourth Medical Center of People's Liberation Army (PLA) General Hospital, Beijing, 100048, China
| | - Keran Song
- Fourth Medical Center of People's Liberation Army (PLA) General Hospital, Beijing, 100048, China
| | - Zhiwei Zhang
- Fourth Medical Center of People's Liberation Army (PLA) General Hospital, Beijing, 100048, China
| | - Junjie Niu
- Department of Orthopaedics, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Lingjun Wang
- Department of Orthopaedics, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Qin Shi
- Department of Orthopaedics, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Liang Chen
- Department of Orthopaedics, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Huilin Yang
- Department of Orthopaedics, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Shuxun Hou
- Fourth Medical Center of People's Liberation Army (PLA) General Hospital, Beijing, 100048, China.
| | - Sai Zhang
- Characteristics Medical Center of the Chinese People's Armed Police Forces (CAPF), Tianjin, 300162, China.
| | - Jianwu Dai
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100190, China.
| |
Collapse
|
30
|
Yin W, Xue W, Zhu H, Shen H, Xiao Z, Wu S, Zhao Y, Cao Y, Tan J, Li J, Liu W, Wang L, Meng L, Chen B, Zhao M, Jiang X, Li X, Ren C, Dai J. Scar tissue removal-activated endogenous neural stem cells aid Taxol-modified collagen scaffolds in repairing chronic long-distance transected spinal cord injury. Biomater Sci 2021; 9:4778-4792. [PMID: 34042920 DOI: 10.1039/d1bm00449b] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Scar tissue removal combined with biomaterial implantation is considered an effective measure to repair chronic transected spinal cord injury (SCI). However, whether more scar tissue removal surgeries could affect the treatment effects of biomaterial implantation still needs to be explored. In this study, we performed the first scar tissue removal surgery in the 3rd month and the second in the 6th month after completely removing 1 cm of spinal tissue in canines. We found that Taxol-modified linear ordered collagen scaffold (LOCS + Taxol) implantation could promote axonal regeneration, neurogenesis, and electrophysiological and functional recovery only in canines at the first scar tissue removal surgery, but not in canines at the second scar tissue removal surgery. Interestingly, we found that more endogenous neural stem cells (NSCs) around the injured site could be activated in canines with the first rather than the second scar tissue removal. Furthermore, we demonstrated that Taxol could promote the neuronal differentiation of NSCs in the myelin inhibition microenvironment through the p38 MAPK signaling pathway in vitro. Therefore, we speculated that endogenous NSC activation by the first scar tissue removal surgery and its further differentiation into neurons induced by Taxol may contribute to functional recovery in canines. Together, LOCS + Taxol implantation in combination with the first scar tissue removal provides a promising therapy for chronic long-distance transected SCI repair with the help of scar tissue removal activated endogenous NSCs.
Collapse
Affiliation(s)
- Wen Yin
- Department of Neurosurgery, Xiangya Hospital, Central South University (CSU), Changsha, Hunan 410008, China.
| | - Weiwei Xue
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Hecheng Zhu
- Changsha Kexin Cancer Hospital, Changsha, Hunan 410205, China
| | - He Shen
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Zhifeng Xiao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Shuyu Wu
- Department of Neurosurgery, Xiangya Hospital, Central South University (CSU), Changsha, Hunan 410008, China.
| | - Yannan Zhao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yudong Cao
- Department of Neurosurgery, Xiangya Hospital, Central South University (CSU), Changsha, Hunan 410008, China.
| | - Jun Tan
- Department of Neurosurgery, Xiangya Hospital, Central South University (CSU), Changsha, Hunan 410008, China.
| | - Juan Li
- Department of Neurosurgery, Xiangya Hospital, Central South University (CSU), Changsha, Hunan 410008, China.
| | - Weidong Liu
- Cancer Research Institute, Key Laboratory for Carcinogenesis of Chinese Ministry of Health, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China.
| | - Lei Wang
- Cancer Research Institute, Key Laboratory for Carcinogenesis of Chinese Ministry of Health, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China.
| | - Li Meng
- Department of Radiology, Xiangya Hospital, Central South University (CSU), Changsha, Hunan 410008, China
| | - Bing Chen
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Ming Zhao
- Changsha Kexin Cancer Hospital, Changsha, Hunan 410205, China
| | - Xingjun Jiang
- Department of Neurosurgery, Xiangya Hospital, Central South University (CSU), Changsha, Hunan 410008, China.
| | - Xing Li
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China and Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Caiping Ren
- Cancer Research Institute, Key Laboratory for Carcinogenesis of Chinese Ministry of Health, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China.
| | - Jianwu Dai
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| |
Collapse
|
31
|
Xue W, Zhang H, Fan Y, Xiao Z, Zhao Y, Liu W, Xu B, Yin Y, Chen B, Li J, Cui Y, Shi Y, Dai J. Upregulation of Apol8 by Epothilone D facilitates the neuronal relay of transplanted NSCs in spinal cord injury. Stem Cell Res Ther 2021; 12:300. [PMID: 34039405 PMCID: PMC8157417 DOI: 10.1186/s13287-021-02375-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 05/09/2021] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Microtubule-stabilizing agents have been demonstrated to modulate axonal sprouting during neuronal disease. One such agent, Epothilone D, has been used to treat spinal cord injury (SCI) by promoting axonal sprouting at the lesion site after SCI. However, the role of Epothilone D in the differentiation of neural stem cells (NSCs) in SCI repair is unknown. In the present study, we mainly explored the effects and mechanisms of Epothilone D on the neuronal differentiation of NSCs and revealed a potential new SCI treatment. METHODS In vitro differentiation assays, western blotting, and quantitative real-time polymerase chain reaction were used to detect the effects of Epothilone D on NSC differentiation. Retrograde tracing using a pseudotyped rabies virus was then used to detect neuronal circuit construction. RNA sequencing (RNA-Seq) was valuable for exploring the target gene involved in the neuronal differentiation stimulated by Epothilone D. In addition, lentivirus-induced overexpression and RNA interference technology were applied to demonstrate the function of the target gene. Last, an Apol8-NSC-linear ordered collagen scaffold (LOCS) graft was prepared to treat a mouse model of SCI, and functional and electrophysiological evaluations were performed. RESULTS We first revealed that Epothilone D promoted the neuronal differentiation of cultured NSCs and facilitated neuronal relay formation in the injured site after SCI. Furthermore, the RNA-Seq results demonstrated that Apol8 was upregulated during Epothilone D-induced neuronal relay formation. Lentivirus-mediated Apol8 overexpression in NSCs (Apol8-NSCs) promoted NSC differentiation toward neurons, and an Apol8 interference assay showed that Apol8 had a role in promoting neuronal differentiation under the induction of Epothilone D. Last, Apol8-NSC transplantation with LOCS promoted the neuronal differentiation of transplanted NSCs in the lesion site as well as synapse formation, thus improving the motor function of mice with complete spinal cord transection. CONCLUSIONS Epothilone D can promote the neuronal differentiation of NSCs by upregulating Apol8, which may provide a promising therapeutic target for SCI repair.
Collapse
Affiliation(s)
- Weiwei Xue
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Haipeng Zhang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.,University of the Chinese Academy of Sciences, Beijing, 100190, China
| | - Yongheng Fan
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.,University of the Chinese Academy of Sciences, Beijing, 100190, China
| | - Zhifeng Xiao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yannan Zhao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Weiyuan Liu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.,University of the Chinese Academy of Sciences, Beijing, 100190, China
| | - Bai Xu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yanyun Yin
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Bing Chen
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jiayin Li
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yi Cui
- Reproductive and Genetic Center of National Research Institute for Family Planning, Beijing, 100081, China
| | - Ya Shi
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jianwu Dai
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China. .,Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China.
| |
Collapse
|
32
|
Liu PC, Tan QW, Zhang Y, Wang H, Zhou L, Yang QR, Xu L, He T, Xie HQ, Lv Q. Hydrogel from acellular porcine adipose tissue promotes survival of adipose tissue transplantation. Biomed Mater 2021; 16. [PMID: 33873165 DOI: 10.1088/1748-605x/abf982] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 04/19/2021] [Indexed: 02/08/2023]
Abstract
Lipofilling is a popular technique for soft tissue augmentation, limited by unpredictable graft survival. This study aimed at exploring the effect of hydrogel from acellular porcine adipose tissue (HAPA) on angiogenesis and survival of adipose tissue used for lipofilling. The effect of HAPA on adipose-derived stem cells (ADSCs) proliferation, adipogenic differentiation, and vascular endothelial growth factor (VEGF) secretion were evaluated in hypoxia and normoxiain vitro. For thein vivostudy, adipose tissue with phosphate buffered saline, ADSCs, and HAPA (with or without ADSCs) were co-injected subcutaneously into nude mice. HAPA-ADSCs mixture (tissue engineering adipose tissue) was also grafted. Gross observation, volume measurement, and ultrasound observation were assessed. For histological assessment, hematoxylin and eosin, perilipin, cluster of differentiation 31 (CD31), Ki67, and transferase-mediated d-UTP nick end labelling (TUNEL) staining were performed. HAPA improved ADSCs proliferation, VEGF secretion, and adipogenic differentiation under normoxia and hypoxia conditionsin vitrostudy. For thein vivostudy, HAPA showed improved volume retention and angiogenesis, and reduced cell apoptosis when compared to ADSCs-assisted lipofilling and pure lipofilling. In conclusion, HAPA could maintain ADSCs viability and improve cell resistant to hypoxia and might be a promising biomaterial to assist lipofilling.
Collapse
Affiliation(s)
- Peng-Cheng Liu
- Department of Breast Surgery, Clinical Research Center for Breast, West China Hospital, Sichuan University, Chengdu, People's Republic of China.,Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Qiu-Wen Tan
- Department of Breast Surgery, Clinical Research Center for Breast, West China Hospital, Sichuan University, Chengdu, People's Republic of China.,Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Yi Zhang
- Research Core Facility of West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Hua Wang
- Department of Breast Surgery, Clinical Research Center for Breast, West China Hospital, Sichuan University, Chengdu, People's Republic of China.,Department of Breast Surgery, Guizhou Provincial People's Hospital, Guiyang, People's Republic of China
| | - Li Zhou
- Research Core Facility of West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Qian-Ru Yang
- Department of Breast Surgery, Clinical Research Center for Breast, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Li Xu
- Department of Breast Surgery, Clinical Research Center for Breast, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Tao He
- Department of Breast Surgery, Clinical Research Center for Breast, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Hui-Qi Xie
- Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Qing Lv
- Department of Breast Surgery, Clinical Research Center for Breast, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| |
Collapse
|
33
|
Collagen-based scaffolds: An auspicious tool to support repair, recovery, and regeneration post spinal cord injury. Int J Pharm 2021; 601:120559. [PMID: 33831486 DOI: 10.1016/j.ijpharm.2021.120559] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/24/2021] [Accepted: 03/27/2021] [Indexed: 02/07/2023]
Abstract
Spinal cord injury (SCI) is a perplexing traumatic disease that habitually gives ride to permanent disability, motor, and sensory impairment. Despite the existence of several therapeutic approaches for the injured motor or sensory neurons, they can't promote axonal regeneration. Whether prepared by conventional or rapid prototyping techniques, scaffolds can be applied to refurbish the continuity of the injured site, by creating a suitable environment for tissue repair, axonal regeneration, and vascularization. Collagen is a multi-sourced protein, found in animals skin, tendons, cartilage, bones, and human placenta, in addition to marine biomass. Collagen is highly abundant in the extracellular matrix and is known for its biocompatibility, biodegradability, porous structure, good permeability, low immunogenicity and thus is extensively applied in the pharmaceutical, cosmetic, and food industries as well as the tissue engineering field. Collagen in scaffolds is usually functionalized with different ligands and factors such as, stem cells, embryonic or human cells to augment its binding specificity and activity. The review summarizes the significance of collagen-based scaffolds and their influence on regeneration, repair and recovery of spinal cord injuries.
Collapse
|
34
|
Lineage tracing reveals the origin of Nestin-positive cells are heterogeneous and rarely from ependymal cells after spinal cord injury. SCIENCE CHINA-LIFE SCIENCES 2021; 65:757-769. [PMID: 33772745 DOI: 10.1007/s11427-020-1901-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 02/22/2021] [Indexed: 12/22/2022]
Abstract
Nestin is expressed extensively in neural stem/progenitor cells during neural development, but its expression is mainly restricted to the ependymal cells in the adult spinal cord. After spinal cord injury (SCI), Nestin expression is reactivated and Nestin-positive (Nestin+) cells aggregate at the injury site. However, the derivation of Nestin+ cells is not clearly defined. Here, we found that Nestin expression was substantially increased in the lesion edge and lesion core after SCI. Using a tamoxifen inducible CreER(T2)-loxP system, we verified that ependymal cells contribute few Nestin+ cells either to the lesion core or the lesion edge after SCI. In the lesion edge, GFAP+ astrocytes were the main cell type that expressed Nestin; they then formed an astrocyte scar. In the lesion core, Nestin+ cells expressed αSMA or Desmin, indicating that they might be derived from pericytes. Our results reveal that Nestin+ cells in the lesion core and edge came from various cell types and rarely from ependymal cells after complete transected SCI, which may provide new insights into SCI repair.
Collapse
|
35
|
Li JJ, Liu H, Zhu Y, Yan L, Liu R, Wang G, Wang B, Zhao B. Animal Models for Treating Spinal Cord Injury Using Biomaterials-Based Tissue Engineering Strategies. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:79-100. [PMID: 33267667 DOI: 10.1089/ten.teb.2020.0267] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Jiao Jiao Li
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Ultimo, Australia
| | - Haifeng Liu
- Department of Orthopedics and Shanxi Medical University Second Affiliated Hospital, Taiyuan, China
| | - Yuanyuan Zhu
- Department of Pharmacy, Shanxi Medical University Second Affiliated Hospital, Taiyuan, China
| | - Lei Yan
- Department of Orthopedics and Shanxi Medical University Second Affiliated Hospital, Taiyuan, China
| | - Ruxing Liu
- Department of Orthopedics and Shanxi Medical University Second Affiliated Hospital, Taiyuan, China
| | - Guishan Wang
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China
| | - Bin Wang
- Department of Orthopedics and Shanxi Medical University Second Affiliated Hospital, Taiyuan, China
- Department of Sports Medicine and Adult Reconstruction Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Bin Zhao
- Department of Orthopedics and Shanxi Medical University Second Affiliated Hospital, Taiyuan, China
| |
Collapse
|
36
|
Alishahi M, Anbiyaiee A, Farzaneh M, Khoshnam SE. Human Mesenchymal Stem Cells for Spinal Cord Injury. Curr Stem Cell Res Ther 2021; 15:340-348. [PMID: 32178619 DOI: 10.2174/1574888x15666200316164051] [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: 06/08/2019] [Revised: 08/03/2019] [Accepted: 09/17/2019] [Indexed: 12/13/2022]
Abstract
Spinal Cord Injury (SCI), as a devastating and life-altering neurological disorder, is one of the most serious health issues. Currently, the management of acute SCI includes pharmacotherapy and surgical decompression. Both the approaches have been observed to have adverse physiological effects on SCI patients. Therefore, novel therapeutic targets for the management of SCI are urgently required for developing cell-based therapies. Multipotent stem cells, as a novel strategy for the treatment of tissue injury, may provide an effective therapeutic option against many neurological disorders. Mesenchymal stem cells (MSCs) or multipotent stromal cells can typically self-renew and generate various cell types. These cells are often isolated from bone marrow (BM-MSCs), adipose tissues (AD-MSCs), umbilical cord blood (UCB-MSCs), and placenta (PMSCs). MSCs have remarkable potential for the development of regenerative therapies in animal models and humans with SCI. Herein, we summarize the therapeutic potential of human MSCs in the treatment of SCI.
Collapse
Affiliation(s)
- Masoumeh Alishahi
- Department of Biology, Tehran North Branch, Islamic Azad University, Tehran, Iran
| | - Amir Anbiyaiee
- Department of Surgery, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 61357-15794, Iran
| | - Maryam Farzaneh
- Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyed E Khoshnam
- Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
37
|
Lewis MJ, Granger N, Jeffery ND. Emerging and Adjunctive Therapies for Spinal Cord Injury Following Acute Canine Intervertebral Disc Herniation. Front Vet Sci 2020; 7:579933. [PMID: 33195591 PMCID: PMC7593405 DOI: 10.3389/fvets.2020.579933] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 09/04/2020] [Indexed: 11/13/2022] Open
Abstract
Some dogs do not make a full recovery following medical or surgical management of acute canine intervertebral disc herniation (IVDH), highlighting the limits of currently available treatment options. The multitude of difficulties in treating severe spinal cord injury are well-recognized, and they have spurred intense laboratory research, resulting in a broad range of strategies that might have value in treating spinal cord-injured dogs. These include interventions that aim to directly repair the spinal cord lesion, promote axonal sparing or regeneration, mitigate secondary injury through neuroprotective mechanisms, or facilitate functional compensation. Despite initial promise in experimental models, many of these techniques have failed or shown mild efficacy in clinical trials in humans and dogs, although high quality evidence is lacking for many of these interventions. However, the continued introduction of new options to the veterinary clinic remains important for expanding our understanding of the mechanisms of injury and repair and for development of novel and combined strategies for severely affected dogs. This review outlines adjunctive or emerging therapies that have been proposed as treatment options for dogs with acute IVDH, including discussion of local or lesion-based approaches as well as systemically applied treatments in both acute and subacute-to-chronic settings. These interventions include low-level laser therapy, electromagnetic fields or oscillating electrical fields, adjunctive surgical techniques (myelotomy or durotomy), systemically or locally-applied hypothermia, neuroprotective chemicals, physical rehabilitation, hyperbaric oxygen therapy, electroacupuncture, electrical stimulation of the spinal cord or specific peripheral nerves, nerve grafting strategies, 4-aminopyridine, chondroitinase ABC, and cell transplantation.
Collapse
Affiliation(s)
- Melissa J Lewis
- Department of Veterinary Clinical Sciences, Purdue University College of Veterinary Medicine, West Lafayette, IN, United States
| | - Nicolas Granger
- The Royal Veterinary College, University of London, Hertfordshire, United Kingdom.,CVS Referrals, Bristol Veterinary Specialists at Highcroft, Bristol, United Kingdom
| | - Nick D Jeffery
- Department of Small Animal Clinical Sciences, Texas A & M College of Veterinary Medicine and Biomedical Sciences, College Station, TX, United States
| | | |
Collapse
|
38
|
Wang Q, He X, Wang B, Pan J, Shi C, Li J, Wang L, Zhao Y, Dai J, Wang D. Injectable collagen scaffold promotes swine myocardial infarction recovery by long-term local retention of transplanted human umbilical cord mesenchymal stem cells. SCIENCE CHINA-LIFE SCIENCES 2020; 64:269-281. [PMID: 32712833 DOI: 10.1007/s11427-019-1575-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 10/25/2019] [Indexed: 12/12/2022]
Abstract
Stem cell therapy is an attractive approach for recovery from myocardial infarction (MI) but faces the challenges of rapid diffusion and poor survival after transplantation. Here we developed an injectable collagen scaffold to promote the long-term retention of transplanted cells in chronic MI. Forty-five minipigs underwent left anterior descending artery (LAD) ligation and were equally divided into three groups 2 months later (collagen scaffold loading with human umbilical mesenchymal stem cell (hUMSC) group, hUMSC group, and placebo group (only phosphate-buffered saline (PBS) injection)). Immunofluorescence staining indicated that the retention of transplanted cells was promoted by the collagen scaffold. Echocardiography and cardiac magnetic resonance imaging (CMR) showed much higher left ventricular ejection fraction (LVEF) and lower infarct size percentage in the collagen/hUMSC group than in the hUMSC and placebo groups at 12 months after treatment. There were also higher densities of vWf-, α-sma-, and cTnT-positive cells in the infarct border zone in the collagen/cell group, as revealed by immunohistochemical analysis, suggesting better angiogenesis and more cardiomyocyte survival after MI. Thus, the injectable collagen scaffold was safe and effective on a large animal myocardial model, which is beneficial for constructing a favorable microenvironment for applying stem cells in clinical MI.
Collapse
Affiliation(s)
- Qiang Wang
- Department of Thoracic and Cardiovascular Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Xiaojun He
- Department of Thoracic and Cardiovascular Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Bin Wang
- Center for Clinical Stem Cell Research, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Jun Pan
- Department of Thoracic and Cardiovascular Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Chunying Shi
- Department of Human Anatomy, Histology and Embryology, School of Basic Medicine, Qingdao University, Qingdao, 266021, China
| | - Jie Li
- Department of Cardiology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Liudi Wang
- Center for Clinical Stem Cell Research, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Yannan Zhao
- Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100190, China.
| | - Jianwu Dai
- Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100190, China.
| | - Dongjin Wang
- Department of Thoracic and Cardiovascular Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China.
| |
Collapse
|
39
|
Vikartovska Z, Kuricova M, Farbakova J, Liptak T, Mudronova D, Humenik F, Madari A, Maloveska M, Sykova E, Cizkova D. Stem Cell Conditioned Medium Treatment for Canine Spinal Cord Injury: Pilot Feasibility Study. Int J Mol Sci 2020; 21:ijms21145129. [PMID: 32698543 PMCID: PMC7404210 DOI: 10.3390/ijms21145129] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/14/2020] [Accepted: 07/14/2020] [Indexed: 12/15/2022] Open
Abstract
Spinal cord injury (SCI) involves nerve damage and often leads to motor, sensory and autonomic dysfunctions. In the present study, we have designed a clinical protocol to assess the feasibility of systemic delivery of allogenic canine bone marrow tissue-derived mesenchymal stem cell conditioned medium (BMMSC CM) to dogs with SCI. Four client-owned dogs with chronic SCI lasting more than six months underwent neurological and clinical evaluation, MRI imaging and blood tests before being enrolled in this study. All dogs received four intravenous infusions with canine allogenic BMMSC CM within one month. Between the infusions the dogs received comprehensive physiotherapy, which continued for three additional months. No adverse effects or complications were observed during the one, three and six months follow-up periods. Neither blood chemistry panel nor hematology profile showed any significant changes. All dogs were clinically improved as assessed using Olby locomotor scales after one, three and six months of BMMSC CM treatment. Furthermore, goniometric measurements revealed partial improvement in the range of joint motion. Bladder function improved in two disabled dogs. We conclude that multiple delivery of allogenic cell-derived conditioned medium to dogs with chronic SCI is feasible, and it might be clinically beneficial in combination with physiotherapy.
Collapse
Affiliation(s)
- Zuzana Vikartovska
- Center of Experimental and Clinical Regenerative Medicine, University of Veterinary Medicine and Pharmacy, Komenskeho 73, 04181 Kosice, Slovakia; (Z.V.); (F.H.); (M.M.)
- University Veterinary Hospital, University of Veterinary Medicine and Pharmacy, Komenskeho 73, 04181 Kosice, Slovakia; (M.K.); (J.F.); (T.L.); (A.M.)
| | - Maria Kuricova
- University Veterinary Hospital, University of Veterinary Medicine and Pharmacy, Komenskeho 73, 04181 Kosice, Slovakia; (M.K.); (J.F.); (T.L.); (A.M.)
| | - Jana Farbakova
- University Veterinary Hospital, University of Veterinary Medicine and Pharmacy, Komenskeho 73, 04181 Kosice, Slovakia; (M.K.); (J.F.); (T.L.); (A.M.)
| | - Tomas Liptak
- University Veterinary Hospital, University of Veterinary Medicine and Pharmacy, Komenskeho 73, 04181 Kosice, Slovakia; (M.K.); (J.F.); (T.L.); (A.M.)
| | - Dagmar Mudronova
- Department of Microbiology and Immunology, Institute of Immunology, University of Veterinary Medicine and Pharmacy, Komenskeho 73, 04181 Kosice, Slovakia;
| | - Filip Humenik
- Center of Experimental and Clinical Regenerative Medicine, University of Veterinary Medicine and Pharmacy, Komenskeho 73, 04181 Kosice, Slovakia; (Z.V.); (F.H.); (M.M.)
| | - Aladar Madari
- University Veterinary Hospital, University of Veterinary Medicine and Pharmacy, Komenskeho 73, 04181 Kosice, Slovakia; (M.K.); (J.F.); (T.L.); (A.M.)
| | - Marcela Maloveska
- Center of Experimental and Clinical Regenerative Medicine, University of Veterinary Medicine and Pharmacy, Komenskeho 73, 04181 Kosice, Slovakia; (Z.V.); (F.H.); (M.M.)
| | - Eva Sykova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, 845 10 Bratislava, Slovakia;
| | - Dasa Cizkova
- Center of Experimental and Clinical Regenerative Medicine, University of Veterinary Medicine and Pharmacy, Komenskeho 73, 04181 Kosice, Slovakia; (Z.V.); (F.H.); (M.M.)
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska cesta 9, 845 10 Bratislava, Slovakia;
- Correspondence:
| |
Collapse
|
40
|
Allotransplantation of adult spinal cord tissues after complete transected spinal cord injury: Long-term survival and functional recovery in canines. SCIENCE CHINA-LIFE SCIENCES 2020; 63:1879-1886. [PMID: 32382980 DOI: 10.1007/s11427-019-1623-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Accepted: 12/05/2019] [Indexed: 12/15/2022]
Abstract
Spinal cord injury (SCI), especially complete transected SCI, leads to loss of cells and extracellular matrix and functional impairments. In a previous study, we transplanted adult spinal cord tissues (aSCTs) to replace lost tissues and facilitate recovery in a rat SCI model. However, rodents display considerable differences from human patients in the scale, anatomy and functions of spinal cord systems, and responses after injury. Thus, use of a large animal SCI model is required to examine the repair efficiency of potential therapeutic approaches. In this study, we transplanted allogenic aSCTs from adult dogs to the lesion area of canines after complete transection of the thoracic spinal cord, and investigated the long-term cell survival and functional recovery. To enhance repair efficiency, a growth factor cocktail was added during aSCT transplantation, providing a favorable microenvironment. The results showed that transplantation of aSCTs, in particular with the addition of growth factors, significantly improves locomotor function restoration and increases the number of neurofilament-, microtubule-associated protein 2-, 5-hydroxytryptamine-, choline acetyltransferase- and tyrosine hydroxylase-positive neurons in the lesion area at 6 months post-surgery. In addition, we demonstrated that donor neurons in aSCTs can survive for a long period after transplantation. This study showed for the first time that transplanting aSCTs combined with growth factor supplementation facilitates reconstruction of injured spinal cords, and consequently promotes long lasting motor function recovery in a large animal complete transected SCI model, and therefore could be considered as a possible therapeutic strategy in humans.
Collapse
|
41
|
Yousefifard M, Nasseri Maleki S, Askarian-Amiri S, Vaccaro AR, Chapman JR, Fehlings MG, Hosseini M, Rahimi-Movaghar V. A combination of mesenchymal stem cells and scaffolds promotes motor functional recovery in spinal cord injury: a systematic review and meta-analysis. J Neurosurg Spine 2020; 32:269-284. [PMID: 31675724 DOI: 10.3171/2019.8.spine19201] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Accepted: 08/01/2019] [Indexed: 11/06/2022]
Abstract
OBJECTIVE There is controversy about the role of scaffolds as an adjunctive therapy to mesenchymal stem cell (MSC) transplantation in spinal cord injury (SCI). Thus, the authors aimed to design a meta-analysis on preclinical evidence to evaluate the effectiveness of combination therapy of scaffold + MSC transplantation in comparison with scaffolds alone and MSCs alone in improving motor dysfunction in SCI. METHODS Electronic databases including Medline, Embase, Scopus, and Web of Science were searched from inception until the end of August 2018. Two independent reviewers screened related experimental studies. Animal studies that evaluated the effectiveness of scaffolds and/or MSCs on motor function recovery following experimental SCI were included. The findings were reported as standardized mean difference (SMD) and 95% confidence interval (CI). RESULTS A total of 34 articles were included in the meta-analysis. Analyses show that combination therapy in comparison with the scaffold group alone (SMD 2.00, 95% CI 1.53-2.46, p < 0.0001), the MSCs alone (SMD 1.58, 95% CI 0.84-2.31, p < 0.0001), and the nontreated group (SMD 3.52, 95% CI 2.84-4.20, p < 0.0001) significantly improved motor function recovery. Co-administration of MSCs + scaffolds only in the acute phase of injury (during the first 3 days after injury) leads to a significant recovery compared to scaffold alone (SMD 2.18, p < 0.0001). In addition, the cotransplantation of scaffolds with bone marrow-derived MSCs (SMD 1.99, p < 0.0001) and umbilical cord-derived MSCs (SMD 1.50, p = 0.001) also improved motor function following SCI. CONCLUSIONS The findings showed that scaffolds + MSCs is more effective than scaffolds and MSCs alone in improving motor function following SCI in animal models, when used in the acute phase of injury.
Collapse
Affiliation(s)
- Mahmoud Yousefifard
- 1Physiology Research Center, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Solmaz Nasseri Maleki
- 1Physiology Research Center, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Alexander R Vaccaro
- 2Department of Orthopedics and Neurosurgery, Rothman Institute, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Jens R Chapman
- 3Swedish Neuroscience Institute, Swedish Medical Center, Seattle, Washington
| | - Michael G Fehlings
- 4Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
- 5Division of Neurosurgery, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
- 6Department of Surgery and Spine Program, University of Toronto, Ontario, Canada
| | - Mostafa Hosseini
- 7Department of Epidemiology and Biostatistics, School of Public Health, and
| | - Vafa Rahimi-Movaghar
- 8Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran; and
- 9Brain and Spinal Injuries Research Center (BASIR), Neuroscience Institute, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
42
|
Deng WS, Ma K, Liang B, Liu XY, Xu HY, Zhang J, Shi HY, Sun HT, Chen XY, Zhang S. Collagen scaffold combined with human umbilical cord-mesenchymal stem cells transplantation for acute complete spinal cord injury. Neural Regen Res 2020; 15:1686-1700. [PMID: 32209773 PMCID: PMC7437585 DOI: 10.4103/1673-5374.276340] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Currently, there is no effective strategy to promote functional recovery after a spinal cord injury. Collagen scaffolds can not only provide support and guidance for axonal regeneration, but can also serve as a bridge for nerve regeneration at the injury site. They can additionally be used as carriers to retain mesenchymal stem cells at the injury site to enhance their effectiveness. Hence, we hypothesized that transplanting human umbilical cord-mesenchymal stem cells on collagen scaffolds would enhance healing following acute complete spinal cord injury. Here, we test this hypothesis through animal studies and a phase I clinical trial. (1) Animal experiments: Models of completely transected spinal cord injury were established in rats and canines by microsurgery. Mesenchymal stem cells derived from neonatal umbilical cord tissue were adsorbed onto collagen scaffolds and surgically implanted at the injury site in rats and canines; the animals were observed after 1 week–6 months. The transplantation resulted in increased motor scores, enhanced amplitude and shortened latency of the motor evoked potential, and reduced injury area as measured by magnetic resonance imaging. (2) Phase I clinical trial: Forty patients with acute complete cervical injuries were enrolled at the Characteristic Medical Center of Chinese People’s Armed Police Force and divided into two groups. The treatment group (n = 20) received collagen scaffolds loaded with mesenchymal stem cells derived from neonatal umbilical cord tissues; the control group (n = 20) did not receive the stem-cell loaded collagen implant. All patients were followed for 12 months. In the treatment group, the American Spinal Injury Association scores and activities of daily life scores were increased, bowel and urinary functions were recovered, and residual urine volume was reduced compared with the pre-treatment baseline. Furthermore, magnetic resonance imaging showed that new nerve fiber connections were formed, and diffusion tensor imaging showed that electrophysiological activity was recovered after the treatment. No serious complication was observed during follow-up. In contrast, the neurological functions of the patients in the control group were not improved over the follow-up period. The above data preliminarily demonstrate that the transplantation of human umbilical cord-mesenchymal stem cells on a collagen scaffold can promote the recovery of neurological function after acute spinal cord injury. In the future, these results need to be confirmed in a multicenter, randomized controlled clinical trial with a larger sample size. The clinical trial was approved by the Ethics Committee of the Characteristic Medical Center of Chinese People’s Armed Police Force on February 3, 2016 (approval No. PJHEC-2016-A8). All animal experiments were approved by the Ethics Committee of the Characteristic Medical Center of Chinese People’s Armed Police Force on May 20, 2015 (approval No. PJHEC-2015-D5).
Collapse
Affiliation(s)
- Wu-Sheng Deng
- College of Integrated Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu Province, China
| | - Ke Ma
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, characteristic medical center of Chinese people's armed police force, Tianjin, China
| | - Bing Liang
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, characteristic medical center of Chinese people's armed police force, Tianjin, China
| | - Xiao-Yin Liu
- Clinical School of Medicine, Tianjin Medical University, Tianjin, China
| | - Hui-You Xu
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, characteristic medical center of Chinese people's armed police force, Tianjin, China
| | - Jian Zhang
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, characteristic medical center of Chinese people's armed police force, Tianjin, China
| | - Heng-Yuan Shi
- Clinical School of Medicine, Logistics University of People's Armed Police Force, Tianjin, China
| | - Hong-Tao Sun
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, characteristic medical center of Chinese people's armed police force, Tianjin, China
| | - Xu-Yi Chen
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, characteristic medical center of Chinese people's armed police force, Tianjin, China
| | - Sai Zhang
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, characteristic medical center of Chinese people's armed police force, Tianjin, China
| |
Collapse
|
43
|
Characterization and immunogenicity of bone marrow-derived mesenchymal stem cells under osteoporotic conditions. SCIENCE CHINA-LIFE SCIENCES 2019; 63:429-442. [PMID: 31879847 DOI: 10.1007/s11427-019-1555-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 09/28/2019] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) are characterized by their multilineage potential and low immunogenicity. However, the properties of MSCs under pathological conditions are unclear. The current study investigated the differentiation potential and immunological characteristics of bone marrow-derived MSCs from ovariectomized-osteoporotic rats (OP-BMSCs). Although the expression of cell morphology- and stemness-related surface markers was similar between OP-BMSCs and BMSCs from healthy rats (H-BMSCs), the proliferation rate was significantly decreased compared with that of H-BMSCs. Regarding multilineage potential, osteogenesis and chondrogenesis abilities of OP-BMSCs decreased, but the adipogenesis ability was significantly enhanced compared with that of H-BMSCs. As expected, decreased osteogenesis following osteogenic induction resulted in reduced expression of β-catenin, osteocalcin, and runt-related transcription factor 2 in OP-BMSCs. Remarkably, the expression of the co-stimulatory proteins CD40 and CD80 was significantly higher, whereas the expression of the negative co-stimulatory molecule programmed cell death ligand 1 was significantly lower in the OP-BMSCs than that in H-BMSCs. Consequently, H-BMSCs inhibited the proliferation and secretion of inflammatory cytokines from anti-CD3 antibody-activated T cells, whereas OP-BMSCs did not. These results indicate that decreased osteogenesis and increased immunogenicity of OP-BMSCs contribute to bone loss in osteoporosis.
Collapse
|
44
|
Cui Y, Yin Y, Xiao Z, Zhao Y, Chen B, Yang B, Xu B, Song H, Zou Y, Ma X, Dai J. LncRNA Neat1 mediates miR-124-induced activation of Wnt/β-catenin signaling in spinal cord neural progenitor cells. Stem Cell Res Ther 2019; 10:400. [PMID: 31852544 PMCID: PMC6921476 DOI: 10.1186/s13287-019-1487-3] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 10/04/2019] [Accepted: 11/05/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Emerging evidence suggests that miR-124 performs important biological functions in neural stem cells (NSCs); it regulates NSC behavior and promotes the differentiation of NSCs into neurons, but the exact molecular mechanism remains unknown. And also, the role of miR-124 during spinal cord injury regeneration is unclear. MATERIALS AND METHODS In order to explore the function of miR-124 in neural differentiation, the molecular markers (Tuj1, Map2, and GFAP) correlated with the differentiation of NSCs were detected by immunofluorescence staining both in cultured mouse spinal cord progenitor cells (SC-NPCs) and in spinal cord injury (SCI) animal models. The migration ability and apoptosis of cultured SC-NPCs were also evaluated by Transwell migration assay and TUNEL assay. In addition, the relative expression of lnRNA Neat1- and Wnt/β-catenin signaling-related genes were detected by quantitative real-time PCR. RESULTS In this study, we revealed that lncRNA Neat1 is involved in regulating Wnt/β-catenin signaling that is activated by miR-124 in SC-NPCs. LncRNA Neat1 was also found to play an important role in regulating neuronal differentiation, apoptosis, and migration of SC-NPCs. Furthermore, we demonstrated that overexpression of miR-124 resulted in elevated Neat1 expression, accompanied with the functional recovery of locomotion in a mouse model of spinal cord injury. CONCLUSIONS Our results confirm the therapeutic effectiveness of miR-124 on the functional recovery of injured spinal cord, supporting the rationale and feasibility of miR-124 for spinal cord injury treatment in future clinical therapy. Furthermore, we concluded that the miR-124-Neat1-Wnt/β-catenin signaling axis is involved in regulating the cell function of SC-NPCs, and this may offer novel therapeutic avenues for future treatment of SCI.
Collapse
Affiliation(s)
- Yi Cui
- Reproductive and Genetic Center of National Research Institute for Family Planning, Beijing, 100081, China
| | - Yanyun Yin
- Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 3 Nanyitiao, Zhongguancun, Beijing, 100080, China
| | - Zhifeng Xiao
- Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 3 Nanyitiao, Zhongguancun, Beijing, 100080, China
| | - Yannan Zhao
- Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 3 Nanyitiao, Zhongguancun, Beijing, 100080, China
| | - Bing Chen
- Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 3 Nanyitiao, Zhongguancun, Beijing, 100080, China
| | - Bin Yang
- Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 3 Nanyitiao, Zhongguancun, Beijing, 100080, China
| | - Bai Xu
- Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 3 Nanyitiao, Zhongguancun, Beijing, 100080, China
| | - Hongwei Song
- Orthopaedics Surgery Department, China-Japan Union Hospital of Jilin University, 126 Xiantai Street, Changchun, 130033, Jilin Province, China
| | - Yunlong Zou
- EHBIO Gene Technology, No. 46, Jiugulou Street, Beijing, 100100, China
| | - Xu Ma
- Reproductive and Genetic Center of National Research Institute for Family Planning, Beijing, 100081, China.
| | - Jianwu Dai
- Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 3 Nanyitiao, Zhongguancun, Beijing, 100080, China.
| |
Collapse
|
45
|
Identification and characterization of two morphologically distinct stem cell subpopulations from human urine samples. SCIENCE CHINA-LIFE SCIENCES 2019; 63:712-723. [PMID: 31515730 DOI: 10.1007/s11427-018-9543-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 05/06/2019] [Indexed: 02/05/2023]
Abstract
Urine-derived stem cells (USCs) have shown potentials for the treatment of skeletal and urological disorders. Based on published literature and our own data, USCs consist of heterogeneous populations of cells. In this paper, we identify and characterize two morphologically distinct subpopulations of USCs from human urine samples, named as spindle-shaped USCs (SS-USCs) and rice-shaped USCs (RS-USCs) respectively. The two subpopulations showed similar clone-forming efficiency, while SS-USCs featured faster proliferation, higher motility, and greater potential for osteogenic and adipogenic differentiation, RS-USCs showed greater potential for chondrogenic differentiation. POU5F1 was strongly expressed in both subpopulations, but MYC was weakly expressed. Both subpopulations showed similar patterns of CD24, CD29, CD34, CD44, CD73, CD90 and CD105 expression, while a higher percentage of RS-USCs were positive for CD133. SS-USCs were positive for VIM, weakly positive for SLC12A1 and UMOD, and negative for KRT18, NPHS1, AQP1 and AQP2, indicating a renal mesenchyme origin; while RS-USCs are positive for VIM, partially positive for KRT18, NPHS1, AQP1, SLC12A1 and UMOD, and negative for AQP2, indicating a nephron tubule origin. The above results can facilitate understanding of the biological characteristics of subpopulations of USCs, and provide a basis for further research and applications of such cells.
Collapse
|
46
|
Pre-Clinical Evaluation of CBD-NT3 Modified Collagen Scaffolds in Completely Spinal Cord Transected Non-Human Primates. J Neurotrauma 2019; 36:2316-2324. [DOI: 10.1089/neu.2018.6078] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
|
47
|
Liu S, Xie YY, Wang B. Role and prospects of regenerative biomaterials in the repair of spinal cord injury. Neural Regen Res 2019; 14:1352-1363. [PMID: 30964053 PMCID: PMC6524500 DOI: 10.4103/1673-5374.253512] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 12/20/2018] [Indexed: 12/12/2022] Open
Abstract
Axonal junction defects and an inhibitory environment after spinal cord injury seriously hinder the regeneration of damaged tissues and neuronal functions. At the site of spinal cord injury, regenerative biomaterials can fill cavities, deliver curative drugs, and provide adsorption sites for transplanted or host cells. Some regenerative biomaterials can also inhibit apoptosis, inflammation and glial scar formation, or further promote neurogenesis, axonal growth and angiogenesis. This review summarized a variety of biomaterial scaffolds made of natural, synthetic, and combined materials applied to spinal cord injury repair. Although these biomaterial scaffolds have shown a certain therapeutic effect in spinal cord injury repair, there are still many problems to be resolved, such as product standards and material safety and effectiveness.
Collapse
Affiliation(s)
- Shuo Liu
- Clinical Stem Cell Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China
| | - Yuan-Yuan Xie
- Clinical Stem Cell Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China
| | - Bin Wang
- Clinical Stem Cell Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China
| |
Collapse
|
48
|
Liu D, Li X, Xiao Z, Yin W, Zhao Y, Tan J, Chen B, Jiang X, Dai J. Different functional bio-scaffolds share similar neurological mechanism to promote locomotor recovery of canines with complete spinal cord injury. Biomaterials 2019; 214:119230. [PMID: 31174066 DOI: 10.1016/j.biomaterials.2019.119230] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 05/22/2019] [Accepted: 05/27/2019] [Indexed: 12/27/2022]
Abstract
Many studies have shown that rodents exhibit a certain degree of spontaneous motor function recovery even if they suffer from spinal cord complete transection injury. However, the characteristics of spontaneous locomotor recovery and its associated neurobiological mechanisms are unclear. In this study, we observed that spontaneous locomotor function recovery of hind limbs could also be detected in a canine thoracic (T8) spinal cord complete transection model. In addition, the spontaneous locomotor recovery of canines could be further promoted by chronic implantation of Taxol- or human bone marrow mesenchymal stem cell-modified bio-scaffolds. Moreover, functional bio-scaffolds implantation promoted locomotor outcome could be significantly weakened (drop to the spontaneous recovery level) but not totally abolished by resection in the lesion site. The neurological mechanism for functional bio-scaffolds improved locomotor outcome was primarily dependent on the formation of neuronal bridging but not the long-distance regeneration of descending motor axons throughout the lesion gap. Besides that, we found that spontaneously achieved locomotor recovery of hind limbs was unable to be weaken by repetitive resection of the lesion area, indicating the mechanism for spontaneous locomotor recovery was independent on functional neurological bridging throughout the lesion gap.
Collapse
Affiliation(s)
- Dingyang Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Xing Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China; Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha 410008, Hunan Province, China; Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China.
| | - Zhifeng Xiao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Wen Yin
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Yannan Zhao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jun Tan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Bing Chen
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xingjun Jiang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China.
| | - Jianwu Dai
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
49
|
Transplantation of adult spinal cord tissue: Transection spinal cord repair and potential clinical translation. SCIENCE CHINA-LIFE SCIENCES 2019; 62:870-872. [PMID: 31124004 DOI: 10.1007/s11427-019-9534-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 04/28/2019] [Accepted: 04/28/2019] [Indexed: 12/22/2022]
|
50
|
Zhang Q, Shi B, Ding J, Yan L, Thawani JP, Fu C, Chen X. Polymer scaffolds facilitate spinal cord injury repair. Acta Biomater 2019; 88:57-77. [PMID: 30710714 DOI: 10.1016/j.actbio.2019.01.056] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 01/10/2019] [Accepted: 01/28/2019] [Indexed: 12/23/2022]
Abstract
During the past decades, improving patient neurological recovery following spinal cord injury (SCI) has remained a challenge. An effective treatment for SCI would not only reduce fractured elements and isolate developing local glial scars to promote axonal regeneration but also ameliorate secondary effects, including inflammation, apoptosis, and necrosis. Three-dimensional (3D) scaffolds provide a platform in which these mechanisms can be addressed in a controlled manner. Polymer scaffolds with favorable biocompatibility and appropriate mechanical properties have been engineered to minimize cicatrization, customize drug release, and ensure an unobstructed space to promote cell growth and differentiation. These properties make polymer scaffolds an important potential therapeutic platform. This review highlights the recent developments in polymer scaffolds for SCI engineering. STATEMENT OF SIGNIFICANCE: How to improve the efficacy of neurological recovery after spinal cord injury (SCI) is always a challenge. Tissue engineering provides a promising strategy for SCI repair, and scaffolds are one of the most important elements in addition to cells and inducing factors. The review highlights recent development and future prospects in polymer scaffolds for SCI therapy. The review will guide future studies by outlining the requirements and characteristics of polymer scaffold technologies employed against SCI. Additionally, the peculiar properties of polymer materials used in the therapeutic process of SCI also have guiding significance to other tissue engineering approaches.
Collapse
|