1
|
Li YX, Zhao LM, Zhang XZ, Ma XK, Liang JQ, Gan TJ, Gong H, Jiang YL, Wu Y, Song YT, Zhang Y, Li Y, Chen XT, Xu CH, Ouyang XY, Li-Ling J, Zhang H, Xie HQ. Smooth muscle extracellular matrix modified small intestinal submucosa conduits promote peripheral nerve repair. Biomaterials 2025; 321:123346. [PMID: 40253732 DOI: 10.1016/j.biomaterials.2025.123346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 04/10/2025] [Accepted: 04/13/2025] [Indexed: 04/22/2025]
Abstract
Challenges still exist to develop an ideal cell-free nerve guidance conduit (NGC) providing a favorable microenvironment for rapid and successful nerve regeneration. Proteomic analysis revealed that extracellular matrix (ECM) derived from smooth muscle cells (SMCs) was abundant in nerve-related active proteins and significantly enriched signaling pathways involved in nerve regeneration. However, whether NGCs based on SMCs-derived ECM modification strategy promote nerve regeneration remains unclear. In the study, we investigated the neuroregenerative effect of SMCs-derived ECM and developed a novel NGC (MyoNerve) by coating small intestinal submucosa (SIS) with SMCs-derived ECM. The SMCs-ECM was rich in neurotrophic factors, which endowed MyoNerve with remarkable neuroregenerative capabilities by promoting the expression of genes implicated in aspects of neuronal maintenance and activating signaling pathways involved in nerve regeneration. In vitro, MyoNerve exhibited excellent bioactivity for accelerating angiogenesis, regulating macrophages polarization, promoting the proliferation, migration and elongation of Schwann cells, enhancing differentiation of PC12 cells, and inducing the neurite outgrowth of dorsal root ganglia. In the model of rat sciatic nerve 10 mm defect, MyoNerve showed great potential for functional nerve regeneration by promoting angiogenesis, proliferation and migration of Schwann cells and neuron, axonal regeneration, remyelination, and neurological functional recovery.
Collapse
Affiliation(s)
- Ya-Xing Li
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Long-Mei Zhao
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xiu-Zhen Zhang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xi-Kun Ma
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University Chengdu, Sichuan, 610041, China
| | - Jing-Qi Liang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Ting-Jiang Gan
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University Chengdu, Sichuan, 610041, China
| | - Heng Gong
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University Chengdu, Sichuan, 610041, China
| | - Yan-Lin Jiang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Ye Wu
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University Chengdu, Sichuan, 610041, China
| | - Yu-Ting Song
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yi Zhang
- Core Facilities of West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yue Li
- Core Facilities of West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiao-Ting Chen
- Animal Experimental Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Cong-Hui Xu
- Department of Radiology, Chengdu Shangjin Nanfu Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiang-Yu Ouyang
- Department of Orthopedics, Hospital of Chengdu Office of People's Government of Xizang Autonomous Region, Chengdu, Sichuan, 610041, China
| | - Jesse Li-Ling
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Hui Zhang
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University Chengdu, Sichuan, 610041, China.
| | - Hui-Qi Xie
- Department of Orthopedic Surgery and Orthopedic Research Institute, Stem Cell and Tissue Engineering Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
2
|
Li X, Zhang J, Zhang Y, Guo L, Gao M, Wang Y, Qiu W, Yuan Y, Zhu J, Liu B, Xiong H, Xu T, Xu R. Conjugated therapy with coaxially printed neural stem cell-laden microfibers and umbilical cord mesenchymal stem cell derived exosomes on complete transactional spinal cord defects. Mater Today Bio 2025; 32:101639. [PMID: 40160243 PMCID: PMC11953994 DOI: 10.1016/j.mtbio.2025.101639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 02/22/2025] [Accepted: 03/03/2025] [Indexed: 04/02/2025] Open
Abstract
Motor function recovery after complete spinal cord injury remained as a challenge in medical field, while one of the key approaches is promoting the local microenvironments. In this research, we performed a conjugated therapy by transplantation of neural stem cell (NSC) scaffolds and umbilical cord mesenchymal stem cell derived exosomes (ucMSC-exos) for the treatment of complete transactional spinal cord injury (SCI). We first demonstrated the anti-inflammatory effects of ucMSC-exos in vitro and found that ucMSC-exos could regulate microglia polarization from M1 to M2, an anti-inflammatory phenotype. Besides, ucMSC-exos also promoted NSC proliferation and neural differentiation during in vitro culturing. On the other hand, core-shell hydrogel microfibers were used as transplantation scaffolds for both small and large SCI defects. The core-shell microfibers could carry large amounts of NSCs in the core portion and the shell portion is highly permeable for nutrient and metabolite transportation. In in vivo experiments, we found that conjugated transplantation of ucMSC-exos and NSC microfibers could decreased inflammatory cytokines at lesion sites, gave rise to more neurons and promoted angiogenesis, thus comprehensively improved the local microenvironment while compared with transplantation of NSC scaffolds only. These beneficial results were in accordance with those in vitro experiments and further led to better locomotor function recovery. In summary, this research has demonstrated that that conjugated transplantation of ucMSC-exos and NSC microfibers could make a potential tool for complete SCI repair.
Collapse
Affiliation(s)
- Xinda Li
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, People's Republic of China
| | - Jin Zhang
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, People's Republic of China
| | - Yi Zhang
- Department of Research and Development, Huaqing Zhimei (Shenzhen) Biotechnology Co., Ltd., Shenzhen, 518107, People's Republic of China
| | - Lili Guo
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, People's Republic of China
| | - Mingjun Gao
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, People's Republic of China
| | - Yangyang Wang
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, People's Republic of China
| | - Wenqiao Qiu
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, People's Republic of China
| | - Ying Yuan
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, People's Republic of China
| | - Jianwei Zhu
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, People's Republic of China
| | - Boxun Liu
- Department of Research and Development, Huaqing Zhimei (Shenzhen) Biotechnology Co., Ltd., Shenzhen, 518107, People's Republic of China
| | - Huan Xiong
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, People's Republic of China
| | - Tao Xu
- Center for Bio-intelligent Manufacturing and Living Matter Bioprinting, Research Institute of Tsinghua University in Shenzhen, Tsinghua University, Shenzhen, 518057, People's Republic of China
| | - Ruxiang Xu
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, People's Republic of China
| |
Collapse
|
3
|
Liu W, Liu Q, Li Z, Zhang C, Li Z, Ke H, Xu X, Wang X, Du H, Talifu Z, Pan Y, Wang X, Mao J, Gao F, Yang D, Yu Y, Liu X, Li J. Multifunctional magneto-electric and exosome-loaded hydrogel enhances neuronal differentiation and immunoregulation through remote non-invasive electrical stimulation for neurological recovery after spinal cord injury. Bioact Mater 2025; 48:510-528. [PMID: 40104021 PMCID: PMC11919302 DOI: 10.1016/j.bioactmat.2025.02.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 02/22/2025] [Accepted: 02/23/2025] [Indexed: 03/20/2025] Open
Abstract
Intervention in the differentiation of neural stem cells (NSCs) is emerging as a highly promising approach for the treatment of spinal cord injury (SCI). However, NSCs at the injury site often suffer from low survival and uncontrolled differentiation. Whereas electrical stimulation has proven effective in regulating the fate of NSCs and promoting tissue repair, however, conventional electrical stimulation therapy has failed to be widely applied due to challenges such as invasiveness and technical complexity. To overcome these limitations, we developed a biomimetic magneto-electric hydrogel incorporating Fe3O4@BaTiO3 core-shell nanoparticles and human umbilical mesenchymal stem cell exosomes (HUMSC-Exos) around the concept of constructing remote noninvasive electrical stimulation for the synergistic treatment of SCI. The Fe3O4@BaTiO3 is activated by the peripheral magnetic field to generate electrical stimulation, which, in conjunction with the synergistic effects of HUMSC-Exos, significantly alleviates the early inflammatory response associated with SCI and enhances the regeneration of newborn neurons and axons, thereby creating favorable conditions for functional recovery post-SCI. Our findings indicate that applying this magneto-exosome hydrogel in a rat model of SCI leads to substantial functional recovery. This innovative combination represents a promising therapeutic strategy for SCI repair.
Collapse
Affiliation(s)
- Wubo Liu
- Department of Orthopaedics, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
- China Rehabilitation Research Center, Beijing Bo'ai Hospital, Beijing, 100068, PR China
- University of Health and Rehabilitation Sciences, Qingdao, Shandong, 266100, PR China
- China Rehabilitation Science Institute, Beijing, 100068, PR China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, 100068, PR China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, PR China
| | - Qiang Liu
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, PR China
| | - Zeqin Li
- China Rehabilitation Research Center, Beijing Bo'ai Hospital, Beijing, 100068, PR China
- China Rehabilitation Science Institute, Beijing, 100068, PR China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, 100068, PR China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, PR China
- Ganan Medical University, Ganzhou, Jiangxi, 341000, PR China
| | - Chunjia Zhang
- China Rehabilitation Research Center, Beijing Bo'ai Hospital, Beijing, 100068, PR China
- China Rehabilitation Science Institute, Beijing, 100068, PR China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, 100068, PR China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, PR China
- School of Rehabilitation, Capital Medical University, Beijing, 100069, PR China
- Department of Rehabilitation Medicine, Peking University Third Hospital, Beijing, 100096, PR China
| | - Zehui Li
- China Rehabilitation Research Center, Beijing Bo'ai Hospital, Beijing, 100068, PR China
- China Rehabilitation Science Institute, Beijing, 100068, PR China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, 100068, PR China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, PR China
- School of Rehabilitation, Capital Medical University, Beijing, 100069, PR China
| | - Han Ke
- Department of Orthopaedics, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
- China Rehabilitation Research Center, Beijing Bo'ai Hospital, Beijing, 100068, PR China
- University of Health and Rehabilitation Sciences, Qingdao, Shandong, 266100, PR China
- China Rehabilitation Science Institute, Beijing, 100068, PR China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, 100068, PR China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, PR China
- Department of Orthopedics, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100013, PR China
| | - Xin Xu
- Department of Orthopaedics, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
- China Rehabilitation Research Center, Beijing Bo'ai Hospital, Beijing, 100068, PR China
- China Rehabilitation Science Institute, Beijing, 100068, PR China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, 100068, PR China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, PR China
- School of Rehabilitation, Capital Medical University, Beijing, 100069, PR China
| | - Xiaoxin Wang
- China Rehabilitation Research Center, Beijing Bo'ai Hospital, Beijing, 100068, PR China
- China Rehabilitation Science Institute, Beijing, 100068, PR China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, 100068, PR China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, PR China
- School of Rehabilitation, Capital Medical University, Beijing, 100069, PR China
| | - Huayong Du
- China Rehabilitation Research Center, Beijing Bo'ai Hospital, Beijing, 100068, PR China
- China Rehabilitation Science Institute, Beijing, 100068, PR China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, 100068, PR China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, PR China
- School of Rehabilitation, Capital Medical University, Beijing, 100069, PR China
| | - Zuliyaer Talifu
- China Rehabilitation Research Center, Beijing Bo'ai Hospital, Beijing, 100068, PR China
- University of Health and Rehabilitation Sciences, Qingdao, Shandong, 266100, PR China
- China Rehabilitation Science Institute, Beijing, 100068, PR China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, 100068, PR China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, PR China
- School of Rehabilitation, Capital Medical University, Beijing, 100069, PR China
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, 100005, PR China
| | - Yunzhu Pan
- China Rehabilitation Research Center, Beijing Bo'ai Hospital, Beijing, 100068, PR China
- University of Health and Rehabilitation Sciences, Qingdao, Shandong, 266100, PR China
- China Rehabilitation Science Institute, Beijing, 100068, PR China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, 100068, PR China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, PR China
- School of Rehabilitation, Capital Medical University, Beijing, 100069, PR China
- Rehabilitation Department, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, 100005, PR China
| | - Xiaoxiong Wang
- Department of Orthopaedics, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
- University of Health and Rehabilitation Sciences, Qingdao, Shandong, 266100, PR China
| | - Jingyun Mao
- College of Environmental and Resource Sciences, College of Carbon Neutral Modern Industry, Fujian Normal University, Fuzhou, 350007, PR China
| | - Feng Gao
- China Rehabilitation Research Center, Beijing Bo'ai Hospital, Beijing, 100068, PR China
- China Rehabilitation Science Institute, Beijing, 100068, PR China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, 100068, PR China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, PR China
- School of Rehabilitation, Capital Medical University, Beijing, 100069, PR China
| | - Degang Yang
- China Rehabilitation Research Center, Beijing Bo'ai Hospital, Beijing, 100068, PR China
- China Rehabilitation Science Institute, Beijing, 100068, PR China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, 100068, PR China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, PR China
- School of Rehabilitation, Capital Medical University, Beijing, 100069, PR China
| | - Yan Yu
- China Rehabilitation Research Center, Beijing Bo'ai Hospital, Beijing, 100068, PR China
- China Rehabilitation Science Institute, Beijing, 100068, PR China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, 100068, PR China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, PR China
- School of Rehabilitation, Capital Medical University, Beijing, 100069, PR China
| | - Xinyu Liu
- Department of Orthopaedics, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
- University of Health and Rehabilitation Sciences, Qingdao, Shandong, 266100, PR China
| | - Jianjun Li
- Department of Orthopaedics, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
- China Rehabilitation Research Center, Beijing Bo'ai Hospital, Beijing, 100068, PR China
- University of Health and Rehabilitation Sciences, Qingdao, Shandong, 266100, PR China
- China Rehabilitation Science Institute, Beijing, 100068, PR China
- Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, 100068, PR China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100068, PR China
- School of Rehabilitation, Capital Medical University, Beijing, 100069, PR China
| |
Collapse
|
4
|
Chen X, Huang R, Yang Z, Zhang J, Yang Y, Gao F, Liu M, Zhang S. Biological engineering approaches for modulating the pathological microenvironment and promoting axonal regeneration after spinal cord injury. Front Neurosci 2025; 19:1574763. [PMID: 40421136 PMCID: PMC12104303 DOI: 10.3389/fnins.2025.1574763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 04/22/2025] [Indexed: 05/28/2025] Open
Abstract
Functional recovery following spinal cord injury (SCI) presents significant challenges and imposes a substantial burden on society. Current research primarily focuses on minimizing damage and promoting regeneration to enhance functional recovery after SCI. Following SCI, secondary injuries such as mitochondrial dysfunction, vascular rupture, inflammatory responses, and glial scarring occur in the lesion area, forming the pathological microenvironment. These factors expand the extent of damage, exacerbate injury severity, and severely impede axonal regeneration after SCI. Modulating the pathological microenvironment through various interventions may facilitate axonal regeneration and promote functional recovery after SCI. This article reviews the influence and research advancements in axon regeneration concerning mitochondrial dysfunction, inflammatory response, and glial scar formation after SCI. Additionally, it integrates insights from bioengineering to improve the pathological microenvironment, summarizing the progress in axon regeneration research. The review concludes with novel strategies for enhancing axon regeneration, offering fresh perspectives for future investigations.
Collapse
Affiliation(s)
- Xiaohong Chen
- Department of Pathology, Yan'an Medical College of Yan'an University, Yan'an, Shaanxi Province, China
| | - Rong Huang
- Department of Pathology, Yan'an Medical College of Yan'an University, Yan'an, Shaanxi Province, China
- Department of General Surgery, Affiliated Hospital of Yan’an University, Yan'an, Shaanxi Province, China
| | - Zhe Yang
- Department of Pathology, Yan'an Medical College of Yan'an University, Yan'an, Shaanxi Province, China
- Department of General Surgery, Affiliated Hospital of Yan’an University, Yan'an, Shaanxi Province, China
| | - Jun Zhang
- Department of Pathology, Yan'an Medical College of Yan'an University, Yan'an, Shaanxi Province, China
- Department of General Surgery, Affiliated Hospital of Yan’an University, Yan'an, Shaanxi Province, China
| | - Yanling Yang
- Department of Pathology, Yan'an Medical College of Yan'an University, Yan'an, Shaanxi Province, China
| | - Feng Gao
- Department of Pathology, Yan'an Medical College of Yan'an University, Yan'an, Shaanxi Province, China
| | - Minli Liu
- Department of Pathology, Yan'an Medical College of Yan'an University, Yan'an, Shaanxi Province, China
| | - Shengjun Zhang
- Department of Pathology, Yan'an Medical College of Yan'an University, Yan'an, Shaanxi Province, China
- Department of General Surgery, Affiliated Hospital of Yan’an University, Yan'an, Shaanxi Province, China
| |
Collapse
|
5
|
Girão AF, Barroca N, Hernández-Martín Y, Completo A, Marques PAAP, Serrano MC. 3D nanofibrous frameworks with on-demand engineered gray and white matters for reconstructing the injured spinal cord. BIOMATERIALS ADVANCES 2025; 170:214200. [PMID: 39904017 DOI: 10.1016/j.bioadv.2025.214200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 12/27/2024] [Accepted: 01/24/2025] [Indexed: 02/06/2025]
Abstract
Spinal cord injury (SCI) is a disruptive and heterogeneous medical condition affecting millions of patients worldwide. Due to the absence of medical treatments to effectively restore the lost sensorimotor and autonomic functions, there is an ongoing pursuit of scaffolds aiming to bridge the injured spinal area. Herein, a novel electrospinning modality to construct 3D nanofibrous frameworks (NFFs) in accordance with distinct spinal cord microenvironments is used to engineer a biomimetic hemicord. This scaffolding concept gravitates around the possibility of customizing NFFs with on-demand engineered gray and white matters to replicate the native spinal cytoarchitecture. In particular, a 3D reduced graphene oxide-based fibrous-porous system is developed to imitate the gray matter, while a 3D polycaprolactone (PCL)-chitosan nanofibrous network combined with PCL-graphene microfibers intends to mimic the white matter. The scaffolding components are tested in vitro with embryonic neural progenitor cells, integrated into the biomimetic NFF, and then tested in vivo in paralyzed rats with cervical hemisection. After 4 months of implantation, the scaffold generates both neuroprotective (e.g., limited infiltration of vimentin+ and ED1+ cells) and neuroregenerative (e.g., presence of new blood vessels and neurites) features accompanied with promising signs of forelimb function recovery.
Collapse
Affiliation(s)
- André F Girão
- Center for Mechanical Technology and Automation (TEMA), Department of Mechanical Engineering, University of Aveiro (UA), Aveiro 3810-193, Portugal; Instituto de Ciencia de Materiales de Madrid (ICMM), Consejo Superior de Investigaciones Científicas (CSIC), Calle Sor Juana Inés de la Cruz 3, Madrid 28049, Spain.
| | - Nathalie Barroca
- Center for Mechanical Technology and Automation (TEMA), Department of Mechanical Engineering, University of Aveiro (UA), Aveiro 3810-193, Portugal
| | - Yasmina Hernández-Martín
- Laboratory of Interfaces for Neural Repair, Hospital Nacional de Parapléjicos, SESCAM, Finca La Peraleda s/n, Toledo 45071, Spain
| | - António Completo
- Center for Mechanical Technology and Automation (TEMA), Department of Mechanical Engineering, University of Aveiro (UA), Aveiro 3810-193, Portugal
| | - Paula A A P Marques
- Center for Mechanical Technology and Automation (TEMA), Department of Mechanical Engineering, University of Aveiro (UA), Aveiro 3810-193, Portugal
| | - María C Serrano
- Instituto de Ciencia de Materiales de Madrid (ICMM), Consejo Superior de Investigaciones Científicas (CSIC), Calle Sor Juana Inés de la Cruz 3, Madrid 28049, Spain.
| |
Collapse
|
6
|
Kalotra S, Kaur G. Neuromethods and assessment tools for traumatic spinal cord injury in rodents: A mini review. Injury 2025; 56:112288. [PMID: 40398195 DOI: 10.1016/j.injury.2025.112288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 02/22/2025] [Accepted: 03/20/2025] [Indexed: 05/23/2025]
Abstract
Spinal cord injury (SCI) is one of the most devastating neurological disorders associated with severe locomotor disability and a high rate of morbidity. Over the last 20-30 years, animal SCI models have proven to be extremely useful in better understanding the underlying molecular mechanism(s) involved in human traumatic SCI and in assessing the efficacy of available therapeutic agents. Thus, the current review article aims to provide readers with an overview of the methods used to induce traumatic SCI and highlight the recent advances in assessment of the functional recovery in rodent models. SCI models are classified into contusion, compression, transection, and Hypoxia-ischemia based on the mechanism of injury caused. Transection injury models are useful for studying the anatomic regeneration and neural circuitries in locomotion, whereas, compression/contusion injury models are used for studying complex biomechanism and neuropathology of human SCI. The ultimate goal of pre-clinical experimental work on traumatic SCI model is to develop effective repair/regenerative strategies for the clinical purpose. Here, we have summarized recent functional recovery assessment tool including quantification of myelin loss and motor neuron counts, axonal regeneration through behavioural and molecular studies.
Collapse
Affiliation(s)
- Shikha Kalotra
- Department of Biotechnology, Guru Nanak Dev University, Amritsar, Punjab 143005, India
| | - Gurcharan Kaur
- Department of Biotechnology, Guru Nanak Dev University, Amritsar, Punjab 143005, India.
| |
Collapse
|
7
|
Cao J, Zhang X, Guo J, Wu J, Lin L, Lin X, Mu J, Huang T, Zhu M, Ma L, Zhou W, Jiang X, Wang X, Feng S, Gu Z, Gao JQ. An engineering-reinforced extracellular vesicle-integrated hydrogel with an ROS-responsive release pattern mitigates spinal cord injury. SCIENCE ADVANCES 2025; 11:eads3398. [PMID: 40173229 PMCID: PMC11963969 DOI: 10.1126/sciadv.ads3398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 02/27/2025] [Indexed: 04/04/2025]
Abstract
The local delivery of mesenchymal stem cell-derived extracellular vesicles (EVs) via hydrogel has emerged as an effective approach for spinal cord injury (SCI) treatment. However, achieving on-demand release of EVs from hydrogel to address dynamically changing pathology remains challenging. Here, we used a series of engineering methods to further enhance EVs' efficacy and optimize their release pattern from hydrogel. Specifically, the pro-angiogenic, neurotrophic, and anti-inflammatory effects of EVs were reinforced through three-dimensional culture and dexamethasone (Dxm) encapsulation. Then, the prepared Dxm-loaded 3EVs (3EVs-Dxm) were membrane modified with ortho-dihydroxy groups (-2OH) and formed an EV-integrated hydrogel (3EVs-Dxm-Gel) via the cross-link with phenylboronic acid-modified hyaluronic acid and tannic acid. The phenylboronic acid ester in 3EVs-Dxm-Gel enabled effective immobilization and reactive oxygen species-responsive release of EVs. Topical injection of 3EVs-Dxm-Gel in SCI rats notably mitigated injury severity and promoted functional recovery, which may offer opportunities for EV-based therapeutics in central nervous system injury.
Collapse
Affiliation(s)
- Jian Cao
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xunqi Zhang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jing Guo
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jiahe Wu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Lingmin Lin
- Department of Rehabilitation Medicine of First Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Xurong Lin
- Department of Rehabilitation Medicine of First Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jiafu Mu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Tianchen Huang
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Manning Zhu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Lan Ma
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Weihang Zhou
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xinchi Jiang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xuhua Wang
- Department of Rehabilitation Medicine of First Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Shiqing Feng
- International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Zhen Gu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Jinhua Institute of Zhejiang University, Jinhua 321002, China
| | - Jian-Qing Gao
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Jinhua Institute of Zhejiang University, Jinhua 321002, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou 310058, China
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| |
Collapse
|
8
|
Wei Y, Zhou X, Li Z, Liu Q, Ding H, Zhou Y, Yin R, Zheng L. Genetically Programmed Single-Component Protein Hydrogel for Spinal Cord Injury Repair. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2405054. [PMID: 39792612 PMCID: PMC11904991 DOI: 10.1002/advs.202405054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 12/02/2024] [Indexed: 01/12/2025]
Abstract
Protein self-assembly allows for the formation of diverse supramolecular materials from relatively simple building blocks. In this study, a single-component self-assembling hydrogel is developed using the recombinant protein CsgA, and its successful application for spinal cord injury repair is demonstrated. Gelation is achieved by the physical entanglement of CsgA nanofibrils, resulting in a self-supporting hydrogel at low concentrations (≥5 mg mL-1). By leveraging the programmability of the CsgA gene sequence, the bioactive hydrogel is enhanced by fusing functional peptide GHK. GHK is recognized for its anti-inflammatory, antioxidant, and neurotrophic factor-stimulating properties, making it a valuable addition to the hydrogel for spinal cord injury repair applications. In vitro experiments demonstrate that the CsgA-GHK hydrogel can modulate microglial M2 polarization, promote neuronal differentiation of neural stem cells, and inhibit astrocyte differentiation. Additionally, the hydrogel shows efficacy in alleviating inflammation and promotes neuronal regeneration at the injury site, leading to significant functional recovery in a rat model with compression injury spinal cord cavity. These findings lay the groundwork for developing a modular design platform for recombinant CsgA protein hydrogels in tissue repair applications.
Collapse
Affiliation(s)
- Yi Wei
- Wenzhou InstituteUniversity of Chinese Academy of SciencesWenzhouZhejiang325001China
| | - Xiaolin Zhou
- Wenzhou InstituteUniversity of Chinese Academy of SciencesWenzhouZhejiang325001China
| | - Zhenhua Li
- Wenzhou InstituteUniversity of Chinese Academy of SciencesWenzhouZhejiang325001China
| | - Qing Liu
- Wenzhou InstituteUniversity of Chinese Academy of SciencesWenzhouZhejiang325001China
| | - Han Ding
- Wenzhou InstituteUniversity of Chinese Academy of SciencesWenzhouZhejiang325001China
| | - Yunlong Zhou
- Wenzhou InstituteUniversity of Chinese Academy of SciencesWenzhouZhejiang325001China
| | - Ruo‐feng Yin
- China‐Japan Union HospitalJilin UniversityChangchunJilin130031China
| | - Lifei Zheng
- Wenzhou InstituteUniversity of Chinese Academy of SciencesWenzhouZhejiang325001China
| |
Collapse
|
9
|
Ma L, Zhang Z, Mu Y, Liu B, Zhou H, Wang DA. The Application of Biomaterial-Based Spinal Cord Tissue Engineering. Macromol Biosci 2025; 25:e2400444. [PMID: 39472074 DOI: 10.1002/mabi.202400444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/11/2024] [Indexed: 03/14/2025]
Abstract
Advancements in biomaterial-based spinal cord tissue engineering technology have profoundly influenced regenerative medicine, providing innovative solutions for both spinal cord organoid development and engineered spinal cord injury (SCI) repair. In spinal cord organoids, biomaterials offer a supportive microenvironment that mimics the natural extracellular matrix, facilitating cell differentiation and organization and advancing the understanding of spinal cord development and pathophysiology. Furthermore, biomaterials are essential in constructing engineered spinal cords for SCI repair. The incorporation of biomaterials with growth factors, fabrication of ordered scaffold structures, and artificial spinal cord assemblies are critical insights for SCI to ensure structural integrity, enhance cell viability, and promote neural regeneration in transplantation. In summary, this review summarizes the contribution of biomaterials to the spinal cord organoids progression and discusses strategies for biomaterial-based spinal cord engineering in SCI therapy. These achievements underscore the transformative potential of biomaterials to improve treatment options for SCI and accelerate future clinical applications.
Collapse
Affiliation(s)
- Liang Ma
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, 999077, China
| | - Zhen Zhang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, 999077, China
| | - Yulei Mu
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, 999077, China
| | - Bangheng Liu
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, 999077, China
| | - Huiqun Zhou
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, 999077, China
| | - Dong-An Wang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, 999077, China
- Centre for Neuromusculoskeletal Restorative Medicine, InnoHK HKSTP, Sha Tin, Hong Kong, 999077, China
- Tung Biomedical Sciences Centre, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, 999077, China
| |
Collapse
|
10
|
Deng M, Xie P, Xue H, Chen Q, Zhou Y, Ming J, Ma Y, Liu J, Huang H. Decellularized tissue matrices hydrogels functionalized with extracellular vesicles promote macrophage reprogramming and neural stem cell differentiation for spinal cord injury repair. J Nanobiotechnology 2025; 23:139. [PMID: 40001048 PMCID: PMC11853540 DOI: 10.1186/s12951-025-03152-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
This study investigates the application of decellularized tissue matrices (DSCM) hydrogels functionalized with extracellular vesicles (EVs) derived from mesenchymal stromal cells (MSCs) for spinal cord injury (SCI) treatment. The primary focus is on how these composites influence macrophage reprogramming and neural stem cell (NSC) differentiation by modulating Slamf9 expression. MSC-derived EVs were successfully isolated, and DSCM hydrogels were prepared from porcine spinal cords. The composite material, EVs derived from MSCs (DSCM@EVs), was constructed and applied to a mouse SCI model, showing significant enhancement in NSC differentiation and axonal growth, thereby alleviating SCI. Bioinformatics and in vitro cell experiments revealed that DSCM@EVs promote the reprogramming of M1 macrophages to the M2 phenotype, reducing inflammatory responses and facilitating NSC differentiation. RNA-seq analysis identified Slamf9 as a key regulatory gene, with its suppression linked to the observed therapeutic effects. This novel approach demonstrates the potential of DSCM@EVs in SCI repair by modulating the inflammatory environment and promoting neural regeneration, offering a promising strategy for treating SCI and potentially other inflammatory neurological disorders.
Collapse
Affiliation(s)
- Ming Deng
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ping Xie
- Department of Chinese Traditional Medicine, Tongren Hospital of Wuhan University (Wuhan Third Hospital), Wuhan, 430060, China
| | - Hongyang Xue
- The First Clinical College of Wuhan University, Wuhan, 430060, China
| | - Qing Chen
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yan Zhou
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jianghua Ming
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yonggang Ma
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Junqi Liu
- Department of Radiation Oncology, The First of Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Hui Huang
- Department of Sports Medicine, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, Hainan Province, 570311, China.
| |
Collapse
|
11
|
Liao Z, Bao Q, Saijilahu, Chimedtseren C, Tumurbaatar K, Saijilafu. Research Progress on Biomaterials for Spinal Cord Repair. Int J Nanomedicine 2025; 20:1773-1787. [PMID: 39958319 PMCID: PMC11829652 DOI: 10.2147/ijn.s501121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 01/22/2025] [Indexed: 02/18/2025] Open
Abstract
Spinal cord injury (SCI) is a very destructive disease of the central nervous system that often causes irreversible nerve damage. Unfortunately, the adult mammalian spinal cord displays little regenerative capacity after injury. In addition, the glial scars and inflammatory responses around the lesion site are another major obstacle for successful axon regeneration after SCI. However, biomaterials are highly biocompatible, and they could provide physical guidance to allow regenerating axon growth over the lesion site and restore functional neural circuits. In addition, combined or synergistic effects of spinal cord repair can be achieved by integrating different strategies, including the use of various biomaterials and microstructures, as well as combining bioactive molecules and living cells. Therefore, it is possible to use tissue engineering scaffolds to regulate the local microenvironment of the injured spinal cord, which may achieve better functional recovery in spinal cord injury repair. In this review, we summarize the latest progress in the treatment of SCI by biomaterials, and discussed its potential mechanism.
Collapse
Affiliation(s)
- Zhenglie Liao
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, People’s Republic of China
| | - Qianyi Bao
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, People’s Republic of China
| | - Saijilahu
- Tongliao Centers for Disease Control and Prevention, Tongliao, Inner Mongolia, People’s Republic of China
| | | | - Khaliunaa Tumurbaatar
- Institute of Traditional Medicine and Technology of Mongolia, Ulaanbaatar city, Mongolia
| | - Saijilafu
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, People’s Republic of China
| |
Collapse
|
12
|
Bradshaw KJ, Leipzig ND. Applications of Regenerative Tissue-Engineered Scaffolds for Treatment of Spinal Cord Injury. Tissue Eng Part A 2025; 31:108-125. [PMID: 39556330 DOI: 10.1089/ten.tea.2024.0194] [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: 11/19/2024] Open
Abstract
Tissue engineering provides a path forward for emerging personalized medicine therapies as well as the ability to bring about cures for diseases or chronic injuries. Traumatic spinal cord injuries (SCIs) are an example of a chronic injury in which no cure or complete functional recovery treatment has been developed. In part, this has been due to the complex and interconnected nature of the central nervous system (CNS), the cellular makeup, its extracellular matrix (ECM), and the injury site pathophysiology. One way to combat the complex nature of an SCI has been to create functional tissue-engineered scaffolds that replace or replenish the aspects of the CNS and tissue/ECM that are damaged following the immediate injury and subsequent immune response. This can be achieved by employing the tissue-engineering triad consisting of cells, biomaterial(s), and environmental factors. Stem cells, with their innate ability to proliferate and differentiate, are a common choice for cellular therapies. Natural or synthetic biomaterials that have tunable characteristics are normally used as the scaffold base. Environmental factors can range from drugs to growth factors (GFs) or proteins, depending on if the idea would be to stimulate exogeneous or endogenous cell populations or just simply retain cells on the scaffold for effective transplantation. For functional regeneration and integration for SCI, the scaffold must promote neuroprotection and neuroplasticity. Tissue-engineering strategies have shown benefits including neuronal differentiation, axonal regeneration, axonal outgrowth, integration into the native spinal cord, and partial functional recovery. Overall, this review focuses on the background that causes SCI to be so difficult to treat, the individual components of the tissue-engineering triad, and how combinatorial scaffolds can be beneficial toward the prospects of future SCI recovery.
Collapse
Affiliation(s)
- Katherine J Bradshaw
- Department of Biomedical Engineering, Auburn Science and Engineering Center #275, The University of Akron, Akron, Ohio, USA
| | - Nic D Leipzig
- Department of Biomedical Engineering, Auburn Science and Engineering Center #275, The University of Akron, Akron, Ohio, USA
- Department of Chemical, Biomolecular, and Corrosion Engineering, The University of Akron, Akron, Ohio, USA
| |
Collapse
|
13
|
Bai J, Liu G, Gao Y, Zhang X, Niu G, Zhang H. Co-culturing neural and bone mesenchymal stem cells in photosensitive hydrogel enhances spinal cord injury repair. Front Bioeng Biotechnol 2024; 12:1431420. [PMID: 39737055 PMCID: PMC11684404 DOI: 10.3389/fbioe.2024.1431420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 11/25/2024] [Indexed: 01/01/2025] Open
Abstract
In mammalian species, neural tissues cannot regenerate following severe spinal cord injury (SCI), for which stem cell transplantation is a promising treatment. Neural stem cells (NSCs) have the potential to repair SCI; however, in unfavourable microenvironments, transplanted NSCs mainly differentiate into astrocytes rather than neurons. In contrast, bone mesenchymal stem cells (BMSCs) promote the differentiation of NSCs into neurons and regulate inflammatory responses. Owing to their easily controllable mechanical properties and similarities to neural tissue, gelatin methacrylate (GelMA) hydrogels offer remarkable cell biocompatibility and regulate the differentiation of NSCs. Therefore, in this study, we propose co-culturing NSCs and BMSCs within low-modulus GelMA hydrogel scaffolds to promote regeneration following SCI. In vitro comparisons revealed that the viability, proliferation, migration, and neuron differentiation capacity of cells in these low-modulus scaffolds exhibit substantially superior performance compared to those in high-modulus hydrogel scaffolds. To the best of our knowledge, this study is the first to report that NSCs/BMSCs co-culture implants can remarkably enhance motor function recovery in SCI rats, reduce the area of spinal cord cavities, stimulate neuron regeneration, and suppress scar tissue formation. Thus, this hydrogel system loaded with co-cultured cells represents a promising therapeutic approach for SCI repair.
Collapse
Affiliation(s)
- Jianzhong Bai
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, Jiangsu, China
- Department of Orthopedics, The Second Affiliated Hospital of Shandong First Medical University, Tai’an, China
| | - Guoping Liu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, Jiangsu, China
- Department of Spine Surgery, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yang Gao
- Department of Orthopedics, The Second Affiliated Hospital of Shandong First Medical University, Tai’an, China
| | - Xishan Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Shandong First Medical University, Tai’an, China
| | - Guoqi Niu
- Department of Orthopedics, The Second Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Hongtao Zhang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
14
|
Li D, Liu S, Lu X, Gong Z, Wang H, Xia X, Lu F, Jiang J, Zhang Y, Xu G, Zou F, Ma X. The Circadian Clock Gene Bmal1 Regulates Microglial Pyroptosis After Spinal Cord Injury via NF-κB/MMP9. CNS Neurosci Ther 2024; 30:e70130. [PMID: 39648661 PMCID: PMC11625957 DOI: 10.1111/cns.70130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 08/07/2024] [Accepted: 10/17/2024] [Indexed: 12/10/2024] Open
Abstract
BACKGROUND The treatment of spinal cord injury (SCI) is usually ineffective, because neuroinflammatory secondary injury is an important cause of the continuous development of spinal cord injury, and microglial pyroptosis is an important step of neuroinflammation. Recently, Bmal1, a core component of circadian clock genes (CCGs), has been shown to play a regulatory role in various tissues and cells. However, it is still unclear whether Bmal1 regulates microglial pyroptosis after SCI. METHODS In this study, we established an in vivo mouse model of SCI using Bmal1 knockout (KO) mice and wild-type (WT) mice, and lipopolysaccharide (LPS)-induced pyroptosis in BV2 cells as an in vitro model. A series of molecular and histological methods were used to detect the level of pyroptosis and explore the regulatory mechanism in vivo and in vitro respectively. RESULTS Both in vitro and in vivo results showed that Bmal1 inhibited NLRP3 inflammasome activation and microglial pyroptosis after SCI. Further analysis showed that Bmal1 inhibited pyroptosis-related proteins (NLRP3, Caspase-1, ASC, GSDMD-N) and reduced the release of IL-18 and IL-1β by inhibiting the NF-κB /MMP9 pathway. It was important that NF-κB was identified as a transcription factor that promotes the expression of MMP9, which in turn regulates microglial pyroptosis after SCI. CONCLUSIONS Our study initially identified that Bmal1 regulates the NF-κB /MMP9 pathway to reduce microglial pyroptosis and thereby reduce secondary spinal cord injury, providing a new promising therapeutic target for SCI.
Collapse
Affiliation(s)
- Dachuan Li
- Department of Orthopedics, Huashan HospitalFudan UniversityShanghaiChina
| | - Siyang Liu
- Department of Orthopedics, Huashan HospitalFudan UniversityShanghaiChina
| | - Xiao Lu
- Department of Orthopedics, Huashan HospitalFudan UniversityShanghaiChina
| | - Zhaoyang Gong
- Department of Orthopedics, Huashan HospitalFudan UniversityShanghaiChina
| | - Hongli Wang
- Department of Orthopedics, Huashan HospitalFudan UniversityShanghaiChina
| | - Xinlei Xia
- Department of Orthopedics, Huashan HospitalFudan UniversityShanghaiChina
| | - Feizhou Lu
- Department of Orthopedics, Huashan HospitalFudan UniversityShanghaiChina
| | - Jianyuan Jiang
- Department of Orthopedics, Huashan HospitalFudan UniversityShanghaiChina
| | - Yuxuan Zhang
- Department of Orthopedics, Huashan HospitalFudan UniversityShanghaiChina
| | - Guangyu Xu
- Department of Orthopedics, Huashan HospitalFudan UniversityShanghaiChina
| | - Fei Zou
- Department of Orthopedics, Huashan HospitalFudan UniversityShanghaiChina
| | - Xiaosheng Ma
- Department of Orthopedics, Huashan HospitalFudan UniversityShanghaiChina
| |
Collapse
|
15
|
Kjar A, Haschert MR, Zepeda JC, Simmons AJ, Yates A, Chavarria D, Fernandez M, Robertson G, Abdulrahman AM, Kim H, Marguerite NT, Moen RK, Drake LE, Curry CW, O'Grady BJ, Gama V, Lau KS, Grueter B, Brunger JM, Lippmann ES. Biofunctionalized gelatin hydrogels support development and maturation of iPSC-derived cortical organoids. Cell Rep 2024; 43:114874. [PMID: 39423129 PMCID: PMC11682736 DOI: 10.1016/j.celrep.2024.114874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 09/16/2024] [Accepted: 09/30/2024] [Indexed: 10/21/2024] Open
Abstract
Human neural organoid models have become an important tool for studying neurobiology. However, improving the representativeness of neural cell populations in such organoids remains a major effort. In this work, we compared Matrigel, a commercially available matrix, to a neural cadherin (N-cadherin) peptide-functionalized gelatin methacryloyl hydrogel (termed GelMA-Cad) for culturing cortical neural organoids. We determined that peptide presentation can tune cell fate and diversity in gelatin-based matrices during differentiation. Of particular note, cortical organoids cultured in GelMA-Cad hydrogels mapped more closely to human fetal populations and produced neurons with more spontaneous excitatory postsynaptic currents relative to Matrigel. These results provide compelling evidence that matrix-tethered signaling peptides can influence neural organoid differentiation, opening an avenue to control stem cell fate. Moreover, outcomes from this work showcase the technical utility of GelMA-Cad as a simple and defined hydrogel alternative to Matrigel for neural organoid culture.
Collapse
Affiliation(s)
- Andrew Kjar
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Mia R Haschert
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - José C Zepeda
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - A Joey Simmons
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA; Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alexis Yates
- Interdisciplinary Materials Science Program, Vanderbilt University, Nashville, TN, USA
| | - Daniel Chavarria
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Melanie Fernandez
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Gabriella Robertson
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Adam M Abdulrahman
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Hyosung Kim
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Nicole T Marguerite
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Rachel K Moen
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Lauren E Drake
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Corinne W Curry
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Brian J O'Grady
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Vivian Gama
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA; Vanderbilt Center for Stem Cell Biology, Vanderbilt University, Nashville, TN, USA
| | - Ken S Lau
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA; Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt Center for Stem Cell Biology, Vanderbilt University, Nashville, TN, USA; Chemical and Physical Biology Program, Vanderbilt University School of Medicine, Nashville, TN, USA; Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Brad Grueter
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA; Department of Anesthesiology, Vanderbilt University, Nashville, TN, USA; Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA
| | - Jonathan M Brunger
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA; Vanderbilt Center for Stem Cell Biology, Vanderbilt University, Nashville, TN, USA; Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ethan S Lippmann
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA; Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA; Interdisciplinary Materials Science Program, Vanderbilt University, Nashville, TN, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA; Vanderbilt Center for Stem Cell Biology, Vanderbilt University, Nashville, TN, USA; Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
16
|
Wei K, Tang C, Ma H, Fang X, Yang R. 3D-printed microrobots for biomedical applications. Biomater Sci 2024; 12:4301-4334. [PMID: 39041236 DOI: 10.1039/d4bm00674g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Microrobots, which can perform tasks in difficult-to-reach parts of the human body under their own or external power supply, are potential tools for biomedical applications, such as drug delivery, microsurgery, imaging and monitoring, tissue engineering, and sensors and actuators. Compared with traditional fabrication methods for microrobots, recent improvements in 3D printers enable them to print high-precision microrobots, breaking through the limitations of traditional micromanufacturing technologies that require high skills for operators and greatly shortening the design-to-production cycle. Here, this review first introduces typical 3D printing technologies used in microrobot manufacturing. Then, the structures of microrobots with different functions and application scenarios are discussed. Next, we summarize the materials (body materials, propulsion materials and intelligent materials) used in 3D microrobot manufacturing to complete body construction and realize biomedical applications (e.g., drug delivery, imaging and monitoring). Finally, the challenges and future prospects of 3D printed microrobots in biomedical applications are discussed in terms of materials, manufacturing and advancement.
Collapse
Affiliation(s)
- Kun Wei
- School of Biomedical Engineering, 3D-Printing and Tissue Engineering Center, Anhui Medical University, Hefei, 230032, China.
| | - Chenlong Tang
- School of Biomedical Engineering, 3D-Printing and Tissue Engineering Center, Anhui Medical University, Hefei, 230032, China.
| | - Hui Ma
- School of Biomedical Engineering, 3D-Printing and Tissue Engineering Center, Anhui Medical University, Hefei, 230032, China.
| | - Xingmiao Fang
- School of Biomedical Engineering, 3D-Printing and Tissue Engineering Center, Anhui Medical University, Hefei, 230032, China.
| | - Runhuai Yang
- School of Biomedical Engineering, 3D-Printing and Tissue Engineering Center, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
17
|
Ju D, Dong C. The combined application of stem cells and three-dimensional bioprinting scaffolds for the repair of spinal cord injury. Neural Regen Res 2024; 19:1751-1758. [PMID: 38103241 PMCID: PMC10960285 DOI: 10.4103/1673-5374.385842] [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: 02/13/2023] [Revised: 06/07/2023] [Accepted: 08/04/2023] [Indexed: 12/18/2023] Open
Abstract
Spinal cord injury is considered one of the most difficult injuries to repair and has one of the worst prognoses for injuries to the nervous system. Following surgery, the poor regenerative capacity of nerve cells and the generation of new scars can make it very difficult for the impaired nervous system to restore its neural functionality. Traditional treatments can only alleviate secondary injuries but cannot fundamentally repair the spinal cord. Consequently, there is a critical need to develop new treatments to promote functional repair after spinal cord injury. Over recent years, there have been several developments in the use of stem cell therapy for the treatment of spinal cord injury. Alongside significant developments in the field of tissue engineering, three-dimensional bioprinting technology has become a hot research topic due to its ability to accurately print complex structures. This led to the loading of three-dimensional bioprinting scaffolds which provided precise cell localization. These three-dimensional bioprinting scaffolds could repair damaged neural circuits and had the potential to repair the damaged spinal cord. In this review, we discuss the mechanisms underlying simple stem cell therapy, the application of different types of stem cells for the treatment of spinal cord injury, and the different manufacturing methods for three-dimensional bioprinting scaffolds. In particular, we focus on the development of three-dimensional bioprinting scaffolds for the treatment of spinal cord injury.
Collapse
Affiliation(s)
- Dingyue Ju
- Department of Anatomy, Medical College of Nantong University, Nantong, Jiangsu Province, China
| | - Chuanming Dong
- Department of Anatomy, Medical College of Nantong University, Nantong, Jiangsu Province, China
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| |
Collapse
|
18
|
Salisbury E, Rawlings TM, Efstathiou S, Tryfonos M, Makwana K, Fitzgerald HC, Gargett CE, Cameron NR, Haddleton DM, Brosens JJ, Eissa AM. Photo-Cross-linked Gelatin Methacryloyl Hydrogels Enable the Growth of Primary Human Endometrial Stromal Cells and Epithelial Gland Organoids. ACS APPLIED MATERIALS & INTERFACES 2024; 16:39140-39152. [PMID: 39022819 PMCID: PMC11299152 DOI: 10.1021/acsami.4c08763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/08/2024] [Accepted: 07/08/2024] [Indexed: 07/20/2024]
Abstract
In vitro three-dimensional (3D) models are better able to replicate the complexity of real organs and tissues than 2D monolayer models. The human endometrium, the inner lining of the uterus, undergoes complex changes during the menstrual cycle and pregnancy. These changes occur in response to steroid hormone fluctuations and elicit crosstalk between the epithelial and stromal cell compartments, and dysregulations are associated with a variety of pregnancy disorders. Despite the importance of the endometrium in embryo implantation and pregnancy establishment, there is a lack of in vitro models that recapitulate tissue structure and function and as such a growing demand for extracellular matrix hydrogels that can support 3D cell culture. To be physiologically relevant, an in vitro model requires mechanical and biochemical cues that mimic those of the ECM found in the native tissue. We report a semisynthetic gelatin methacryloyl (GelMA) hydrogel that combines the bioactive properties of natural hydrogels with the tunability and reproducibility of synthetic materials. We then describe a simple protocol whereby cells can quickly be encapsulated in GelMA hydrogels. We investigate the suitability of GelMA hydrogel to support the development of an endometrial model by culturing the main endometrial cell types: stromal cells and epithelial cells. We also demonstrate how the mechanical and biochemical properties of GelMA hydrogels can be tailored to support the growth and maintenance of epithelial gland organoids that emerge upon 3D culturing of primary endometrial epithelial progenitor cells in a defined chemical medium. We furthermore demonstrate the ability of GelMA hydrogels to support the viability of stromal cells and their function measured by monitoring decidualization in response to steroid hormones. This study describes the first steps toward the development of a hydrogel matrix-based model that recapitulates the structure and function of the native endometrium and could support applications in understanding reproductive failure.
Collapse
Affiliation(s)
- Emma Salisbury
- Department
of Chemistry, University of Warwick, Coventry CV4 7AL, U.K.
| | - Thomas M. Rawlings
- Division
of Biomedical Sciences, Reproductive Health Unit, Clinical Science
Research Laboratories, Warwick Medical School, University of Warwick
and Tommy’s National Centre for Miscarriage Research, University
Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, U.K.
| | | | - Maria Tryfonos
- Division
of Biomedical Sciences, Reproductive Health Unit, Clinical Science
Research Laboratories, Warwick Medical School, University of Warwick
and Tommy’s National Centre for Miscarriage Research, University
Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, U.K.
| | - Komal Makwana
- Division
of Biomedical Sciences, Reproductive Health Unit, Clinical Science
Research Laboratories, Warwick Medical School, University of Warwick
and Tommy’s National Centre for Miscarriage Research, University
Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, U.K.
| | - Harriet C. Fitzgerald
- The
Ritchie Centre, Hudson Institute of Medical Research, Clayton VIC 3168, Australia
- Department
of Obstetrics and Gynaecology, Monash University, Clayton VIC 3168, Australia
| | - Caroline E. Gargett
- The
Ritchie Centre, Hudson Institute of Medical Research, Clayton VIC 3168, Australia
- Department
of Obstetrics and Gynaecology, Monash University, Clayton VIC 3168, Australia
| | - Neil R. Cameron
- Department
of Materials Science and Engineering, Monash
University, Clayton, Victoria 3800, Australia
- School of
Engineering, University of Warwick, Coventry CV4 7AL, U.K.
| | | | - Jan J. Brosens
- Division
of Biomedical Sciences, Reproductive Health Unit, Clinical Science
Research Laboratories, Warwick Medical School, University of Warwick
and Tommy’s National Centre for Miscarriage Research, University
Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, U.K.
| | - Ahmed M. Eissa
- Department
of Chemistry, University of Warwick, Coventry CV4 7AL, U.K.
- Department
of Polymers, Chemical Industries Research Division, National Research
Centre, El Bohouth St.
33, Dokki, Cairo Giza 12622, Egypt
- School
of Life Sciences, Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton WV1 1LY, U.K.
| |
Collapse
|
19
|
Li C, Luo Y, Li S. The roles of neural stem cells in myelin regeneration and repair therapy after spinal cord injury. Stem Cell Res Ther 2024; 15:204. [PMID: 38978125 PMCID: PMC11232222 DOI: 10.1186/s13287-024-03825-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 07/02/2024] [Indexed: 07/10/2024] Open
Abstract
Spinal cord injury (SCI) is a complex tissue injury that results in a wide range of physical deficits, including permanent or progressive disabilities of sensory, motor and autonomic functions. To date, limitations in current clinical treatment options can leave SCI patients with lifelong disabilities. There is an urgent need to develop new therapies for reconstructing the damaged spinal cord neuron-glia network and restoring connectivity with the supraspinal pathways. Neural stem cells (NSCs) possess the ability to self-renew and differentiate into neurons and neuroglia, including oligodendrocytes, which are cells responsible for the formation and maintenance of the myelin sheath and the regeneration of demyelinated axons. For these properties, NSCs are considered to be a promising cell source for rebuilding damaged neural circuits and promoting myelin regeneration. Over the past decade, transplantation of NSCs has been extensively tested in a variety of preclinical models of SCI. This review aims to highlight the pathophysiology of SCI and promote the understanding of the role of NSCs in SCI repair therapy and the current advances in pathological mechanism, pre-clinical studies, as well as clinical trials of SCI via NSC transplantation therapeutic strategy. Understanding and mastering these frontier updates will pave the way for establishing novel therapeutic strategies to improve the quality of recovery from SCI.
Collapse
Affiliation(s)
- Chun Li
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Neurology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, Tongji University School of Medicine, Shanghai, 200092, China
| | - Yuping Luo
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Neurology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Siguang Li
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Neurology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China.
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China.
| |
Collapse
|
20
|
Li R, Feng J, Li L, Luo G, Shi Y, Shen S, Yuan X, Wu J, Yan B, Yang L. Recombinant fibroblast growth factor 4 ameliorates axonal regeneration and functional recovery in acute spinal cord injury through altering microglia/macrophage phenotype. Int Immunopharmacol 2024; 134:112188. [PMID: 38728880 DOI: 10.1016/j.intimp.2024.112188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 04/18/2024] [Accepted: 04/28/2024] [Indexed: 05/12/2024]
Abstract
Neuroinflammation is one of the extensive secondary injury processes that aggravate metabolic and cellular dysfunction and tissue loss following spinal cord injury (SCI). Thus, an anti-inflammatory strategy is crucial for modulating structural and functional restoration during the stage of acute and chronic SCI. Recombinant fibroblast growth factor 4 (rFGF4) has eliminated its mitogenic activity and demonstrated a metabolic regulator for alleviating hyperglycemia in type 2 diabetes and liver injury in non-alcoholic steatohepatitis. However, it remains to be explored whether or not rFGF4 has a neuroprotective effect for restoring neurological disorders, such as SCI. Here, we identified that rFGF4 could polarize microglia/macrophages into the restorative M2 subtype, thus exerting an anti-inflammatory effect to promote neurological functional recovery and nerve fiber regeneration after SCI. Importantly, these effects by rFGF4 were related to triggering PI3K/AKT/GSK3β and attenuating TLR4/NF-κB signaling axes. Conversely, gene silencing of the PI3K/AKT/GSK3β signaling or pharmacological reactivation of the TLR4/NF-κB axis aggravated inflammatory reaction. Thus, our findings highlight rFGF4 as a potentially therapeutic regulator for repairing SCI, and its outstanding effect is associated with regulating macrophage/microglial polarization.
Collapse
Affiliation(s)
- Rui Li
- Orthopaedics/Department of Spine Surgery, Department of Pharmacy and Department of Gastroenterology, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine), Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, China; State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Juerong Feng
- Orthopaedics/Department of Spine Surgery, Department of Pharmacy and Department of Gastroenterology, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine), Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, China
| | - Liuxun Li
- Orthopaedics/Department of Spine Surgery, Department of Pharmacy and Department of Gastroenterology, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine), Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, China
| | - Guotian Luo
- Orthopaedics/Department of Spine Surgery, Department of Pharmacy and Department of Gastroenterology, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine), Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, China
| | - Yongpeng Shi
- Orthopaedics/Department of Spine Surgery, Department of Pharmacy and Department of Gastroenterology, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine), Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, China
| | - Shichao Shen
- Orthopaedics/Department of Spine Surgery, Department of Pharmacy and Department of Gastroenterology, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine), Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, China
| | - Xinrong Yuan
- Orthopaedics/Department of Spine Surgery, Department of Pharmacy and Department of Gastroenterology, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine), Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, China
| | - Jianlong Wu
- Orthopaedics/Department of Spine Surgery, Department of Pharmacy and Department of Gastroenterology, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine), Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, China
| | - Bin Yan
- Orthopaedics/Department of Spine Surgery, Department of Pharmacy and Department of Gastroenterology, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine), Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, China.
| | - Lei Yang
- Orthopaedics/Department of Spine Surgery, Department of Pharmacy and Department of Gastroenterology, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine), Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, China.
| |
Collapse
|
21
|
Lee HW, Chen KT, Li YCE, Yeh YC, Chiang CY, Lee IC. Dual crosslinking silk fibroin/pectin-based bioink development and the application on neural stem/progenitor cells spheroid laden 3D bioprinting. Int J Biol Macromol 2024; 269:131720. [PMID: 38677692 DOI: 10.1016/j.ijbiomac.2024.131720] [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: 10/16/2023] [Revised: 01/14/2024] [Accepted: 04/18/2024] [Indexed: 04/29/2024]
Abstract
The human nervous system is an incredibly intricate physiological network, and neural cells lack the ability to repair and regenerate after a brain injury. 3-dimensional (3D) bioprinting technology offers a promising strategy for constructing biomimetic organ constructs and in vitro brain/disease models. The bioink serves as a pivotal component that emulates the microenvironment of biomimetic construct and exerts a profound influence on cellular behaviors. In this study, a series of mechanically adjustable and dual crosslinking bioinks were developed using photocrosslinkable methacrylated silk fibroin (SilMA) in combination with the ionic crosslinking material, pectin, or pectin methacryloyl (PecMA) with silk fibroin (SF) supplementation. SilMA/pectin exhibited superior properties, with SilMA providing biocompatibility and adjustable mechanical properties, while the addition of pectin enhanced printability. The porous structure supported neural cell growth, and 15 % SilMA/0.5 % pectin bioinks displayed excellent printability and shape fidelity. Neural stem/progenitor cells (NSPCs)-loaded bioinks were used to construct a 3D brain model, demonstrating sustained vitality and high neuronal differentiation without the need for growth factors. The SilMA/pectin bioinks demonstrated adjustable mechanical properties, favorable biocompatibility, and an environment highly conducive to neural induction, offering an alternative approach for neural tissue engineering applications or in vitro brain models.
Collapse
Affiliation(s)
- Hao-Wei Lee
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Ko-Ting Chen
- Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Yi-Chen Ethan Li
- Department of Chemical Engineering, Feng Chia University, Taichung 40724, Taiwan
| | - Yu-Chun Yeh
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Chao-Ying Chiang
- Department of Chemical Engineering, Feng Chia University, Taichung 40724, Taiwan
| | - I-Chi Lee
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
22
|
Li X, Li Y, Zhang X, Xu J, Kang J, Li B, Zhao B, Wang L. Cross-Linking Methods of the Silk Protein Hydrogel in Oral and Craniomaxillofacial Tissue Regeneration. Tissue Eng Regen Med 2024; 21:529-544. [PMID: 38294593 PMCID: PMC11087422 DOI: 10.1007/s13770-023-00624-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 12/16/2023] [Accepted: 12/17/2023] [Indexed: 02/01/2024] Open
Abstract
BACKGROUND Craniomaxillofacial tissue defects are clinical defects involving craniomaxillofacial and oral soft and hard tissues. They are characterized by defect-shaped irregularities, bacterial and inflammatory environments, and the need for functional recovery. Conventional clinical treatments are currently unable to achieve regeneration of high-quality oral craniomaxillofacial tissue. As a natural biomaterial, silk fibroin (SF) has been widely studied in biomedicine and has broad prospects for use in tissue regeneration. Hydrogels made of SF showed excellent water retention, biocompatibility, safety and the ability to combine with other materials. METHODS To gain an in-depth understanding of the current development of SF, this article reviews the structure, preparation and application prospects in oral and craniomaxillofacial tissue regenerative medicine. It first briefly introduces the structure of SF and then summarizes the principles, advantages and disadvantages of the different cross-linking methods (physical cross-linking, chemical cross-linking and double network structure) of SF. Finally, the existing research on the use of SF in tissue engineering and the prospects of using SF with different cross-linking methods in oral and craniomaxillofacial tissue regeneration are also discussed. CONCLUSIONS This review is intended to show the advantages of SF hydrogels in tissue engineering and provides theoretical support for establishing novel and viable silk protein hydrogels for regeneration.
Collapse
Affiliation(s)
- Xiujuan Li
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China
| | - Yuanjiao Li
- School of Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xinsong Zhang
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China
| | - Jie Xu
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China
| | - Jie Kang
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China
| | - Bing Li
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China
| | - Bin Zhao
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China.
| | - Lu Wang
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China.
| |
Collapse
|
23
|
Vinnacombe-Willson GA, García-Astrain C, Troncoso-Afonso L, Wagner M, Langer J, González-Callejo P, Silvio DD, Liz-Marzán LM. Growing Gold Nanostars on 3D Hydrogel Surfaces. CHEMISTRY OF MATERIALS : A PUBLICATION OF THE AMERICAN CHEMICAL SOCIETY 2024; 36:5192-5203. [PMID: 38828187 PMCID: PMC11137816 DOI: 10.1021/acs.chemmater.4c00564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/11/2024] [Accepted: 04/12/2024] [Indexed: 06/05/2024]
Abstract
Nanocomposites comprising hydrogels and plasmonic nanoparticles are attractive materials for tissue engineering, bioimaging, and biosensing. These materials are usually fabricated by adding colloidal nanoparticles to the uncured polymer mixture and thus require time-consuming presynthesis, purification, and ligand-exchange steps. Herein, we introduce approaches for rapid synthesis of gold nanostars (AuNSt) in situ on hydrogel substrates, including those with complex three-dimensional (3D) features. These methods enable selective AuNSt growth at the surface of the substrate, and the growth conditions can be tuned to tailor the nanoparticle size and density (coverage). We additionally demonstrate proof-of-concept applications of these nanocomposites for SERS sensing and imaging. High surface coverage with AuNSt enabled 1-2 orders of magnitude higher SERS signals compared to plasmonic hydrogels loaded with premade colloids. Importantly, AuNSt can be prepared without the addition of any potentially cytotoxic surfactants, thereby ensuring a high biocompatibility. Overall, in situ growth becomes a versatile and straightforward approach for the fabrication of plasmonic biomaterials.
Collapse
Affiliation(s)
| | - Clara García-Astrain
- CIC
biomaGUNE, Basque Research and Technology
Alliance (BRTA), Donostia-San
Sebastián 20014, Spain
- Centro
de Investigación Biomédica en Red de Bioingeniería
Biomateriales, y Nanomedicina (CIBER-BBN), Donostia-San Sebastián 20014, Spain
| | - Lara Troncoso-Afonso
- CIC
biomaGUNE, Basque Research and Technology
Alliance (BRTA), Donostia-San
Sebastián 20014, Spain
- Department
of Applied Chemistry, University of the
Basque Country (UPV-EHU), Donostia-San
Sebastián 20018, Spain
| | - Marita Wagner
- CIC
biomaGUNE, Basque Research and Technology
Alliance (BRTA), Donostia-San
Sebastián 20014, Spain
- Department
of Applied Chemistry, University of the
Basque Country (UPV-EHU), Donostia-San
Sebastián 20018, Spain
- CIC
nanoGUNE, Basque Research and Technology
Alliance (BRTA), Donostia-San Sebastián 20018, Spain
| | - Judith Langer
- CIC
biomaGUNE, Basque Research and Technology
Alliance (BRTA), Donostia-San
Sebastián 20014, Spain
| | | | - Desirè Di Silvio
- CIC
biomaGUNE, Basque Research and Technology
Alliance (BRTA), Donostia-San
Sebastián 20014, Spain
| | - Luis M. Liz-Marzán
- CIC
biomaGUNE, Basque Research and Technology
Alliance (BRTA), Donostia-San
Sebastián 20014, Spain
- Centro
de Investigación Biomédica en Red de Bioingeniería
Biomateriales, y Nanomedicina (CIBER-BBN), Donostia-San Sebastián 20014, Spain
- Ikerbasque
Basque Foundation for Science, Bilbao 48009, Spain
- Cinbio, Universidade de Vigo, Vigo 36310, Spain
| |
Collapse
|
24
|
Deng B, Jiang S, Liu G, Li X, Zhao Y, Fan X, Ren J, Ning C, Xu L, Ji L, Mu X. Tetramethylpyrazine-loaded electroconductive hydrogels promote tissue repair after spinal cord injury by protecting the blood-spinal cord barrier and neurons. J Mater Chem B 2024; 12:4409-4426. [PMID: 38630533 DOI: 10.1039/d3tb02160b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2024]
Abstract
Spinal cord injury (SCI) usually induces profound microvascular dysfunction. It disrupts the integrity of the blood-spinal cord barrier (BSCB), which could trigger a cascade of secondary pathological events that manifest as neuronal apoptosis and axonal demyelination. These events can further lead to irreversible neurological impairments. Thus, reducing the permeability of the BSCB and maintaining its substructural integrity are essential to promote neuronal survival following SCI. Tetramethylpyrazine (TMP) has emerged as a potential protective agent for treating the BSCB after SCI. However, its therapeutic potential is hindered by challenges in the administration route and suboptimal bioavailability, leading to attenuated clinical outcomes. To address this challenge, traditional Chinese medicine, TMP, was used in this study to construct a drug-loaded electroconductive hydrogel for synergistic treatment of SCI. A conductive hydrogel combined with TMP demonstrates good electrical and mechanical properties as well as superior biocompatibility. Furthermore, it also facilitates sustained local release of TMP at the implantation site. Furthermore, the TMP-loaded electroconductive hydrogel could suppress oxidative stress responses, thereby diminishing endothelial cell apoptosis and the breakdown of tight junction proteins. This concerted action repairs BSCB integrity. Concurrently, myelin-associated axons and neurons are protected against death, which meaningfully restore neurological functions post spinal cord injury. Hence, these findings indicate that combining the electroconductive hydrogel with TMP presents a promising avenue for potentiating drug efficacy and synergistic repair following SCI.
Collapse
Affiliation(s)
- Bowen Deng
- Division of Intelligent and Biomechanical System, State Key Laboratory of Tribology, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China.
- Department of Orthopedics, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China.
| | - Shengyuan Jiang
- Department of Orthopedics, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China.
| | - Gang Liu
- Department of Orthopedics, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China.
| | - Xiaoye Li
- Department of Orthopedics, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China.
| | - Yi Zhao
- Department of Orthopedics, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China.
| | - Xiao Fan
- Department of Orthopedics, Qingdao Municipal Hospital, Qingdao, 266071, Shandong Province, China
| | - Jingpei Ren
- Department of Orthopedics, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China.
| | - Chengyun Ning
- College of Materials Science and Technology, South China University of Technology, Guangzhou 510641, Guangdong Province, China
| | - Lin Xu
- Department of Orthopedics, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China.
| | - Linhong Ji
- Division of Intelligent and Biomechanical System, State Key Laboratory of Tribology, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China.
| | - Xiaohong Mu
- Department of Orthopedics, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China.
| |
Collapse
|
25
|
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
|
26
|
Song P, Han T, Wu Z, Fang H, Liu Y, Ying W, Wang X, Shen C. Transplantation of Neural Stem Cells Loaded in an IGF-1 Bioactive Supramolecular Nanofiber Hydrogel for the Effective Treatment of Spinal Cord Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306577. [PMID: 38441409 PMCID: PMC11077690 DOI: 10.1002/advs.202306577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 01/11/2024] [Indexed: 05/09/2024]
Abstract
Spinal cord injury (SCI) leads to massive cell death, disruption, and demyelination of axons, resulting in permanent motor and sensory dysfunctions. Stem cell transplantation is a promising therapy for SCI. However, owing to the poor microenvironment that develops following SCI, the bioactivities of these grafted stem cells are limited. Cell implantation combined with biomaterial therapies is widely studied for the development of tissue engineering technology. Herein, an insulin-like growth factor-1 (IGF-1)-bioactive supramolecular nanofiber hydrogel (IGF-1 gel) is synthesized that can activate IGF-1 downstream signaling, prevent the apoptosis of neural stem cells (NSCs), improve their proliferation, and induce their differentiation into neurons and oligodendrocytes. Moreover, implantation of NSCs carried out with IGF-1 gels promotes neurite outgrowth and myelin sheath regeneration at lesion sites following SCI. In addition, IGF-1 gels can enrich extracellular vesicles (EVs) derived from NSCs or from nerve cells differentiated from these NSCs via miRNAs related to axonal regeneration and remyelination, even in an inflammatory environment. These EVs are taken up by autologous endogenous NSCs and regulate their differentiation. This study provides adequate evidence that combined treatment with NSCs and IGF-1 gels is a potential therapeutic strategy for treating SCI.
Collapse
Affiliation(s)
- Peiwen Song
- Department of Orthopedics (Spinal Surgery)Laboratory of Spinal and Spinal Cord Injury Regeneration and RepairThe First Affiliated Hospital of Anhui Medical UniversityHefei230032China
- Anhui Province Research Center for the Clinical Application of Medical TechnologyThe First Affiliated Hospital of Anhui Medical UniversityHefei230032China
| | - Tianyu Han
- Department of Orthopedics (Spinal Surgery)Laboratory of Spinal and Spinal Cord Injury Regeneration and RepairThe First Affiliated Hospital of Anhui Medical UniversityHefei230032China
- Anhui Province Research Center for the Clinical Application of Medical TechnologyThe First Affiliated Hospital of Anhui Medical UniversityHefei230032China
| | - Zuomeng Wu
- Department of Orthopedics (Spinal Surgery)Laboratory of Spinal and Spinal Cord Injury Regeneration and RepairThe First Affiliated Hospital of Anhui Medical UniversityHefei230032China
- Anhui Province Research Center for the Clinical Application of Medical TechnologyThe First Affiliated Hospital of Anhui Medical UniversityHefei230032China
| | - Huang Fang
- Department of Orthopedics (Spinal Surgery)The First Affiliated Hospital of USTCHefei230032China
| | - Yunlei Liu
- Department of Clinical LaboratoryThe First Affiliated Hospital of Anhui Medical UniversityHefei230032China
| | - Wang Ying
- Department of Medical ImagingThe First Affiliated Hospital of Anhui Medical UniversityHefei230032China
| | - Xianwen Wang
- School of Biomedical EngineeringResearch and Engineering Center of Biomedical MaterialsAnhui Provincial Institute of Translational MedicineAnhui Medical UniversityHefei230032P. R. China
| | - Cailiang Shen
- Department of Orthopedics (Spinal Surgery)Laboratory of Spinal and Spinal Cord Injury Regeneration and RepairThe First Affiliated Hospital of Anhui Medical UniversityHefei230032China
- Anhui Province Research Center for the Clinical Application of Medical TechnologyThe First Affiliated Hospital of Anhui Medical UniversityHefei230032China
| |
Collapse
|
27
|
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
|
28
|
Das S, Jegadeesan JT, Basu B. Gelatin Methacryloyl (GelMA)-Based Biomaterial Inks: Process Science for 3D/4D Printing and Current Status. Biomacromolecules 2024; 25:2156-2221. [PMID: 38507816 DOI: 10.1021/acs.biomac.3c01271] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
Tissue engineering for injured tissue replacement and regeneration has been a subject of investigation over the last 30 years, and there has been considerable interest in using additive manufacturing to achieve these goals. Despite such efforts, many key questions remain unanswered, particularly in the area of biomaterial selection for these applications as well as quantitative understanding of the process science. The strategic utilization of biological macromolecules provides a versatile approach to meet diverse requirements in 3D printing, such as printability, buildability, and biocompatibility. These molecules play a pivotal role in both physical and chemical cross-linking processes throughout the biofabrication, contributing significantly to the overall success of the 3D printing process. Among the several bioprintable materials, gelatin methacryloyl (GelMA) has been widely utilized for diverse tissue engineering applications, with some degree of success. In this context, this review will discuss the key bioengineering approaches to identify the gelation and cross-linking strategies that are appropriate to control the rheology, printability, and buildability of biomaterial inks. This review will focus on the GelMA as the structural (scaffold) biomaterial for different tissues and as a potential carrier vehicle for the transport of living cells as well as their maintenance and viability in the physiological system. Recognizing the importance of printability toward shape fidelity and biophysical properties, a major focus in this review has been to discuss the qualitative and quantitative impact of the key factors, including microrheological, viscoelastic, gelation, shear thinning properties of biomaterial inks, and printing parameters, in particular, reference to 3D extrusion printing of GelMA-based biomaterial inks. Specifically, we emphasize the different possibilities to regulate mechanical, swelling, biodegradation, and cellular functionalities of GelMA-based bio(material) inks, by hybridization techniques, including different synthetic and natural biopolymers, inorganic nanofillers, and microcarriers. At the close, the potential possibility of the integration of experimental data sets and artificial intelligence/machine learning approaches is emphasized to predict the printability, shape fidelity, or biophysical properties of GelMA bio(material) inks for clinically relevant tissues.
Collapse
Affiliation(s)
- Soumitra Das
- Materials Research Centre, Indian Institute of Science, Bangalore, India 560012
| | | | - Bikramjit Basu
- Materials Research Centre, Indian Institute of Science, Bangalore, India 560012
| |
Collapse
|
29
|
Hosseini SM, Borys B, Karimi-Abdolrezaee S. Neural stem cell therapies for spinal cord injury repair: an update on recent preclinical and clinical advances. Brain 2024; 147:766-793. [PMID: 37975820 DOI: 10.1093/brain/awad392] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 10/22/2023] [Accepted: 11/02/2023] [Indexed: 11/19/2023] Open
Abstract
Traumatic spinal cord injury (SCI) is a leading cause of lifelong disabilities. Permanent sensory, motor and autonomic impairments after SCI are substantially attributed to degeneration of spinal cord neurons and axons, and disintegration of neural network. To date, minimal regenerative treatments are available for SCI with an unmet need for new therapies to reconstruct the damaged spinal cord neuron-glia network and restore connectivity with the supraspinal pathways. Multipotent neural precursor cells (NPCs) have a unique capacity to generate neurons, oligodendrocytes and astrocytes. Due to this capacity, NPCs have been an attractive cell source for cellular therapies for SCI. Transplantation of NPCs has been extensively tested in preclinical models of SCI in the past two decades. These studies have identified opportunities and challenges associated with NPC therapies. While NPCs have the potential to promote neuroregeneration through various mechanisms, their low long-term survival and integration within the host injured spinal cord limit the functional benefits of NPC-based therapies for SCI. To address this challenge, combinatorial strategies have been developed to optimize the outcomes of NPC therapies by enriching SCI microenvironment through biomaterials, genetic and pharmacological therapies. In this review, we will provide an in-depth discussion on recent advances in preclinical NPC-based therapies for SCI. We will discuss modes of actions and mechanism by which engrafted NPCs contribute to the repair process and functional recovery. We will also provide an update on current clinical trials and new technologies that have facilitated preparation of medical-grade human NPCs suitable for transplantation in clinical studies.
Collapse
Affiliation(s)
- Seyed Mojtaba Hosseini
- Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Rady Faculty of Health Sciences, University of Manitoba Winnipeg, Manitoba R3E 0J9, Canada
- Manitoba Multiple Sclerosis Research Center, Winnipeg, Manitoba R3E 0J9, Canada
| | - Ben Borys
- Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Rady Faculty of Health Sciences, University of Manitoba Winnipeg, Manitoba R3E 0J9, Canada
| | - Soheila Karimi-Abdolrezaee
- Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Rady Faculty of Health Sciences, University of Manitoba Winnipeg, Manitoba R3E 0J9, Canada
- Manitoba Multiple Sclerosis Research Center, Winnipeg, Manitoba R3E 0J9, Canada
- Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba R3E 3P4, Canada
| |
Collapse
|
30
|
Gao H, Liu S, Qin S, Yang J, Yue T, Ye B, Tang Y, Feng J, Hou J, Danzeng D. Injectable hydrogel-based combination therapy for myocardial infarction: a systematic review and Meta-analysis of preclinical trials. BMC Cardiovasc Disord 2024; 24:119. [PMID: 38383333 PMCID: PMC10882925 DOI: 10.1186/s12872-024-03742-0] [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: 09/30/2023] [Accepted: 01/19/2024] [Indexed: 02/23/2024] Open
Abstract
INTRODUCTION This study evaluates the effectiveness of a combined regimen involving injectable hydrogels for the treatment of experimental myocardial infarction. PATIENT CONCERNS Myocardial infarction is an acute illness that negatively affects quality of life and increases mortality rates. Experimental models of myocardial infarction can aid in disease research by allowing for the development of therapies that effectively manage disease progression and promote tissue repair. DIAGNOSIS Experimental animal models of myocardial infarction were established using the ligation method on the anterior descending branch of the left coronary artery (LAD). INTERVENTIONS The efficacy of intracardiac injection of hydrogels, combined with cells, drugs, cytokines, extracellular vesicles, or nucleic acid therapies, was evaluated to assess the functional and morphological improvements in the post-infarction heart achieved through the combined hydrogel regimen. OUTCOMES A literature review was conducted using PubMed, Web of Science, Scopus, and Cochrane databases. A total of 83 papers, including studies on 1332 experimental animals (rats, mice, rabbits, sheep, and pigs), were included in the meta-analysis based on the inclusion and exclusion criteria. The overall effect size observed in the group receiving combined hydrogel therapy, compared to the group receiving hydrogel treatment alone, resulted in an ejection fraction (EF) improvement of 8.87% [95% confidence interval (CI): 7.53, 10.21] and a fractional shortening (FS) improvement of 6.31% [95% CI: 5.94, 6.67] in rat models, while in mice models, the improvements were 16.45% [95% CI: 11.29, 21.61] for EF and 5.68% [95% CI: 5.15, 6.22] for FS. The most significant improvements in EF (rats: MD = 9.63% [95% CI: 4.02, 15.23]; mice: MD = 23.93% [95% CI: 17.52, 30.84]) and FS (rats: MD = 8.55% [95% CI: 2.54, 14.56]; mice: MD = 5.68% [95% CI: 5.15, 6.22]) were observed when extracellular vesicle therapy was used. Although there have been significant results in large animal experiments, the number of studies conducted in this area is limited. CONCLUSION The present study demonstrates that combining hydrogel with other therapies effectively improves heart function and morphology. Further preclinical research using large animal models is necessary for additional study and validation.
Collapse
Affiliation(s)
- Han Gao
- School of Medicine, Tibet University, Lhasa, Tibet, China
| | - Song Liu
- School of Medicine, Tibet University, Lhasa, Tibet, China
| | - Shanshan Qin
- School of Medicine, Tibet University, Lhasa, Tibet, China
| | - Jiali Yang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Tian Yue
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Bengui Ye
- West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China
| | - Yue Tang
- School of Pharmacy, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Jie Feng
- School of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Jun Hou
- Department of Cardiology, Chengdu Third People's Hospital, Chengdu, Sichuan, China.
| | - Dunzhu Danzeng
- School of Medicine, Tibet University, Lhasa, Tibet, China.
| |
Collapse
|
31
|
Yang Y, Ma B, Chen J, Liu D, Ma J, Li B, Hao J, Zhou X. Epigenetic regulation and factors that influence the effect of iPSCs-derived neural stem/progenitor cells (NS/PCs) in the treatment of spinal cord injury. Clin Epigenetics 2024; 16:30. [PMID: 38383473 PMCID: PMC10880347 DOI: 10.1186/s13148-024-01639-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 01/30/2024] [Indexed: 02/23/2024] Open
Abstract
Spinal cord injury (SCI) is a severe neurological disorder that causes neurological impairment and disability. Neural stem/progenitor cells (NS/PCs) derived from induced pluripotent stem cells (iPSCs) represent a promising cell therapy strategy for spinal cord regeneration and repair. However, iPSC-derived NS/PCs face many challenges and issues in SCI therapy; one of the most significant challenges is epigenetic regulation and that factors that influence this mechanism. Epigenetics refers to the regulation of gene expression and function by DNA methylation, histone modification, and chromatin structure without changing the DNA sequence. Previous research has shown that epigenetics plays a crucial role in the generation, differentiation, and transplantation of iPSCs, and can influence the quality, safety, and outcome of transplanted cells. In this study, we review the effects of epigenetic regulation and various influencing factors on the role of iPSC-derived NS/PCs in SCI therapy at multiple levels, including epigenetic reprogramming, regulation, and the adaptation of iPSCs during generation, differentiation, and transplantation, as well as the impact of other therapeutic tools (e.g., drugs, electrical stimulation, and scaffolds) on the epigenetic status of transplanted cells. We summarize our main findings and insights in this field and identify future challenges and directions that need to be addressed and explored.
Collapse
Affiliation(s)
- Yubiao Yang
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Boyuan Ma
- The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, People's Republic of China
| | - Jinyu Chen
- The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, People's Republic of China
| | - Derong Liu
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Jun Ma
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Bo Li
- Department of Orthopedics, Beijing Luhe Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Jian Hao
- The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, People's Republic of China.
| | - Xianhu Zhou
- The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, People's Republic of China.
| |
Collapse
|
32
|
Chapla R, Katz RR, West JL. Neurogenic Cell Behavior in 3D Culture Enhanced Within a Highly Compliant Synthetic Hydrogel Platform Formed via Competitive Crosslinking. Cell Mol Bioeng 2024; 17:35-48. [PMID: 38435792 PMCID: PMC10901766 DOI: 10.1007/s12195-024-00794-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 01/09/2024] [Indexed: 03/05/2024] Open
Abstract
Purpose Scaffold materials that better support neurogenesis are still needed to improve cell therapy outcomes for neural tissue damage. We have used a modularly tunable, highly compliant, degradable hydrogel to explore the impacts of hydrogel compliance stiffness on neural differentiation. Here we implemented competitive matrix crosslinking mechanics to finely tune synthetic hydrogel moduli within soft tissue stiffnesses, a range much softer than typically achievable in synthetic crosslinked hydrogels, providing a modularly controlled and ultrasoft 3D culture model which supports and enhances neurogenic cell behavior. Methods Soluble competitive allyl monomers were mixed with proteolytically-degradable poly(ethylene glycol) diacrylate derivatives and crosslinked to form a matrix, and resultant hydrogel stiffness and diffusive properties were evaluated. Neural PC12 cells or primary rat fetal neural stem cells (NSCs) were encapsulated within the hydrogels, and cell morphology and phenotype were investigated to understand cell-matrix interactions and the effects of environmental stiffness on neural cell behavior within this model. Results Addition of allyl monomers caused a concentration-dependent decrease in hydrogel compressive modulus from 4.40 kPa to 0.26 kPa (natural neural tissue stiffness) without influencing soluble protein diffusion kinetics through the gel matrix. PC12 cells encapsulated in the softest hydrogels showed significantly enhanced neurite extension in comparison to PC12s in all other hydrogel stiffnesses tested. Encapsulated neural stem cells demonstrated significantly greater spreading and elongation in 0.26 kPa alloc hydrogels than in 4.4 kPa hydrogels. When soluble growth factor deprivation (for promotion of neural differentiation) was evaluated within the neural stiffness gels (0.26 kPa), NSCs showed increased neuronal marker expression, indicating early enhancement of neurogenic differentiation. Conclusions Implementing allyl-acrylate crosslinking competition reduced synthetic hydrogel stiffness to provide a supportive environment for 3D neural tissue culture, resulting in enhanced neurogenic behavior of encapsulated cells. These results indicate the potential suitability of this ultrasoft hydrogel system as a model platform for further investigating environmental factors on neural cell behavior. Supplementary Information The online version contains supplementary material available at 10.1007/s12195-024-00794-2.
Collapse
Affiliation(s)
- Rachel Chapla
- Department of Biomedical Engineering, Duke University, Durham, NC 27708 USA
| | - Rachel R. Katz
- Department of Biomedical Engineering, Duke University, Durham, NC 27708 USA
| | - Jennifer L. West
- Department of Biomedical Engineering, Duke University, Durham, NC 27708 USA
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22904 USA
| |
Collapse
|
33
|
Gao Y, Wang K, Wu S, Wu J, Zhang J, Li J, Lei S, Duan X, Men K. Injectable and Photocurable Gene Scaffold Facilitates Efficient Repair of Spinal Cord Injury. ACS APPLIED MATERIALS & INTERFACES 2024; 16:4375-4394. [PMID: 38185858 DOI: 10.1021/acsami.3c14902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
RNA interference-based gene therapy has led to a strategy for spinal cord injury (SCI) therapy. However, there have been high requirements regarding the optimal gene delivery vector for siRNA-based SCI gene therapy. Here, we developed an injectable and photocurable lipid nanoparticle GelMA (PLNG) hydrogel scaffold for controlled dual siRNA delivery at the SCI wound site. The prepared PLNG scaffold could efficiently protect and retain the bioactivity of the siRNA nanocomplex. It facilitated sustainable siRNA release along with degradation in 7 days. After loading dual siRNA targeting phosphatase and tensin homologue (PTEN) and macrophage migration inhibitory factor (MIF) simultaneously, the locally administered siRNAs/PLNG scaffold efficiently improved the Basso mouse scale (BMS) score and recovered ankle joint movement and plantar stepping after treatment with only three doses. We further proved that the siRNAs/PLNG scaffold successfully regulated the activities of neurons, microglia, and macrophages, thus promoting neuron axon regeneration and remyelination. The protein array results suggested that the siRNAs/PLNG scaffold could increase the expression of growth factors and decrease the expression of inflammatory factors to regulate neuroinflammation in SCI and create a neural repair environment. Our results suggested that the PLNG scaffold siRNA delivery system is a potential candidate for siRNA-based SCI therapy.
Collapse
Affiliation(s)
- Yan Gao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Kaiyu Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shan Wu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jieping Wu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jin Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jingmei Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Sibei Lei
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xingmei Duan
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Ke Men
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
34
|
Ozulumba T, Zatorski JM, Arneja A, Hammel JH, Braciale TJ, Luckey CJ, Munson JM, Pompano RR. Mitigating reactive oxygen species production and increasing gel porosity improves lymphocyte motility and fibroblast spreading in photocrosslinked gelatin-thiol hydrogels. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.14.574282. [PMID: 38293038 PMCID: PMC10827049 DOI: 10.1101/2024.01.14.574282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
On-chip 3D culture systems that incorporate immune cells such as lymphocytes and stromal cells are needed to model immune organs in engineered systems such as organs-on-chip. Photocrosslinking is a useful tool for creating such immune-competent hydrogel cultures with spatial cell organization. However, loss of viability and motility in photocrosslinked gels can limit its utility, especially when working with fragile primary cells. We hypothesized that optimizing photoexposure-induced ROS production, hydrogel porosity or a combination of both factors was necessary to sustain cell viability and motility during culture in photocrosslinked gelatin-thiol (GelSH) hydrogels. Jurkat T cells, primary human CD4+ T cells and human lymphatic fibroblasts were selected as representative lymphoid immune cells to test this hypothesis. Direct exposure of these cells to 385 nm light and LAP photoinitiator dramatically increased ROS levels. Pretreatment with an antioxidant, ascorbic acid (AA), protected the cells from light + LAP-induced ROS and was non-toxic at optimized doses. Furthermore, scanning electron microscopy showed that native GelSH hydrogels had limited porosity, and that adding collagen to GelSH precursor before crosslinking markedly increased gel porosity. Next, we tested the impact of AA pretreatment and increasing gel porosity, alone or in combination, on cell viability and function in 3D GelSH hydrogel cultures. Increasing gel porosity, rather than AA pretreatment, was more critical for rescuing viability of Jurkat T cells and spreading of human lymphatic fibroblasts in GelSH-based gels, but both factors improved the motility of primary human CD4+ T cells. Increased porosity enabled formation of spatially organized co-cultures of primary human CD4+ T cells and human lymphatic fibroblasts in photo-crosslinked gels in a multi-lane microfluidic chip, towards modeling the lymphoid organ microenvironment. Some optimization is still needed to improve homogeneity between regions on the chip. These findings will enable researchers utilizing photocrosslinking methods to develop immunocompetent 3D culture models that support viability and function of sensitive lymphoid cells.
Collapse
|
35
|
Giorgi Z, Veneruso V, Petillo E, Veglianese P, Perale G, Rossi F. Biomaterials and Cell Therapy Combination in Central Nervous System Treatments. ACS APPLIED BIO MATERIALS 2024; 7:80-98. [PMID: 38158393 PMCID: PMC10792669 DOI: 10.1021/acsabm.3c01058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/07/2023] [Accepted: 12/11/2023] [Indexed: 01/03/2024]
Abstract
Current pharmacological and surgical therapies for the central nervous system (CNS) show a limited capacity to reduce the damage progression; that together with the intrinsic limited capability of the CNS to regenerate greatly reduces the hopes of recovery. Among all the therapies proposed, the tissue engineering strategies supplemented with therapeutic stem cells remain the most promising. Neural tissue engineering strategies are based on the development of devices presenting optimal physical, chemical, and mechanical properties which, once inserted in the injured site, can support therapeutic cells, limiting the effect of a hostile environment and supporting regenerative processes. Thus, this review focuses on the employment of hydrogel and nanofibrous scaffolds supplemented with stem cells as promising therapeutic tools for the central and peripheral nervous systems in preclinical and clinical applications.
Collapse
Affiliation(s)
- Zoe Giorgi
- Department
of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, piazza Leonardo da Vinci 32, 20133, Milan, Italy
| | - Valeria Veneruso
- Istituto
di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
- Faculty
of Biomedical Sciences, University of Southern
Switzerland (USI), Via
Buffi 13, 6900 Lugano, Switzerland
| | - Emilia Petillo
- Department
of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, piazza Leonardo da Vinci 32, 20133, Milan, Italy
- Istituto
di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
| | - Pietro Veglianese
- Istituto
di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
- Faculty
of Biomedical Sciences, University of Southern
Switzerland (USI), Via
Buffi 13, 6900 Lugano, Switzerland
| | - Giuseppe Perale
- Faculty
of Biomedical Sciences, University of Southern
Switzerland (USI), Via
Buffi 13, 6900 Lugano, Switzerland
- Ludwig
Boltzmann Institute for Experimental and Clinical Traumatology, Donaueschingenstrasse 13, 1200 Vienna, Austria
| | - Filippo Rossi
- Department
of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, piazza Leonardo da Vinci 32, 20133, Milan, Italy
- Faculty
of Biomedical Sciences, University of Southern
Switzerland (USI), Via
Buffi 13, 6900 Lugano, Switzerland
| |
Collapse
|
36
|
Guan P, Fan L, Zhu Z, Yang Q, Kang X, Li J, Zhang Z, Liu S, Liu C, Wang X, Xu J, Wang K, Sun Y. M2 microglia-derived exosome-loaded electroconductive hydrogel for enhancing neurological recovery after spinal cord injury. J Nanobiotechnology 2024; 22:8. [PMID: 38167113 PMCID: PMC10763283 DOI: 10.1186/s12951-023-02255-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 12/06/2023] [Indexed: 01/05/2024] Open
Abstract
Electroconductive hydrogels offer a promising avenue for enhancing the repair efficacy of spinal cord injuries (SCI) by restoring disrupted electrical signals along the spinal cord's conduction pathway. Nonetheless, the application of hydrogels composed of diverse electroconductive materials has demonstrated limited capacity to mitigate the post-SCI inflammatory response. Recent research has indicated that the transplantation of M2 microglia effectively fosters SCI recovery by attenuating the excessive inflammatory response. Exosomes (Exos), small vesicles discharged by cells carrying similar biological functions to their originating cells, present a compelling alternative to cellular transplantation. This investigation endeavors to exploit M2 microglia-derived exosomes (M2-Exos) successfully isolated and reversibly bonded to electroconductive hydrogels through hydrogen bonding for synergistic promotion of SCI repair to synergistically enhance SCI repair. In vitro experiments substantiated the significant capacity of M2-Exos-laden electroconductive hydrogels to stimulate the growth of neural stem cells and axons in the dorsal root ganglion and modulate microglial M2 polarization. Furthermore, M2-Exos demonstrated a remarkable ability to mitigate the initial inflammatory reaction within the injury site. When combined with the electroconductive hydrogel, M2-Exos worked synergistically to expedite neuronal and axonal regeneration, substantially enhancing the functional recovery of rats afflicted with SCI. These findings underscore the potential of M2-Exos as a valuable reparative factor, amplifying the efficacy of electroconductive hydrogels in their capacity to foster SCI rehabilitation.
Collapse
Affiliation(s)
- Pengfei Guan
- Department of Pediatric Orthopedic, Center for Orthopedic Surgery, the Third Affiliated Hospital of Southern Medical University, Guangzhou, 510515, China
| | - Lei Fan
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Zhaobo Zhu
- Department of Orthopedic Surgery, Nanfang Hospital Baiyun Branch, Southern Medical University, Guangzhou, 510080, China
| | - Qinfeng Yang
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xinchang Kang
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Junji Li
- Department of Pediatric Orthopedic, Center for Orthopedic Surgery, the Third Affiliated Hospital of Southern Medical University, Guangzhou, 510515, China
| | - Zuyu Zhang
- Department of Pediatric Orthopedic, Center for Orthopedic Surgery, the Third Affiliated Hospital of Southern Medical University, Guangzhou, 510515, China
| | - Shencai Liu
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Can Liu
- Department of Spine Surgery, Center for Orthopedic Surgery, the Third Affiliated Hospital of Southern Medical University, Guangzhou, 510515, China
| | - Xuelian Wang
- The Operating Room of the Third Affiliated Hospital of Southern Medical University, Guangzhou, 510515, China
| | - Jing Xu
- Department of Ophthalmology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Kun Wang
- Department of Spine Surgery, Department of Orthopedics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| | - Yongjian Sun
- Department of Pediatric Orthopedic, Center for Orthopedic Surgery, the Third Affiliated Hospital of Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
37
|
Marques-Almeida T, Lanceros-Mendez S, Ribeiro C. State of the Art and Current Challenges on Electroactive Biomaterials and Strategies for Neural Tissue Regeneration. Adv Healthc Mater 2024; 13:e2301494. [PMID: 37843074 DOI: 10.1002/adhm.202301494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 09/22/2023] [Indexed: 10/17/2023]
Abstract
The loss or failure of an organ/tissue stands as one of the healthcare system's most prevalent, devastating, and costly challenges. Strategies for neural tissue repair and regeneration have received significant attention due to their particularly strong impact on patients' well-being. Many research efforts are dedicated not only to control the disease symptoms but also to find solutions to repair the damaged tissues. Neural tissue engineering (TE) plays a key role in addressing this problem and significant efforts are being carried out to develop strategies for neural repair treatment. In the last years, active materials allowing to tune cell-materials interaction are being increasingly used, representing a recent paradigm in TE applications. Among the most important stimuli influencing cell behavior are the electrical and mechanical ones. In this way, materials with the ability to provide this kind of stimuli to the neural cells seem to be appropriate to support neural TE. In this scope, this review summarizes the different biomaterials types used for neural TE, highlighting the relevance of using active biomaterials and electrical stimulation. Furthermore, this review provides not only a compilation of the most relevant studies and results but also strategies for novel and more biomimetic approaches for neural TE.
Collapse
Affiliation(s)
- Teresa Marques-Almeida
- Physics Centre of Minho and Porto Universities (CF-UM-UP), University of Minho, Braga, 4710-057, Portugal
- LaPMET - Laboratory of Physics for Materials and Emergent Technologies, University of Minho, Braga, 4710-057, Portugal
| | - Senentxu Lanceros-Mendez
- Physics Centre of Minho and Porto Universities (CF-UM-UP), University of Minho, Braga, 4710-057, Portugal
- LaPMET - Laboratory of Physics for Materials and Emergent Technologies, University of Minho, Braga, 4710-057, Portugal
- BCMaterials, Basque Center for Materials, Applications and Nanostructures, UPV/EHU Science Park, Leioa, 48940, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, 48009, Spain
| | - Clarisse Ribeiro
- Physics Centre of Minho and Porto Universities (CF-UM-UP), University of Minho, Braga, 4710-057, Portugal
- LaPMET - Laboratory of Physics for Materials and Emergent Technologies, University of Minho, Braga, 4710-057, Portugal
| |
Collapse
|
38
|
Lisboa ES, Serafim C, Santana W, Dos Santos VLS, de Albuquerque-Junior RLC, Chaud MV, Cardoso JC, Jain S, Severino P, Souto EB. Nanomaterials-combined methacrylated gelatin hydrogels (GelMA) for cardiac tissue constructs. J Control Release 2024; 365:617-639. [PMID: 38043727 DOI: 10.1016/j.jconrel.2023.11.056] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 11/21/2023] [Accepted: 11/28/2023] [Indexed: 12/05/2023]
Abstract
Among non-communicable diseases, cardiovascular diseases are the most prevalent, accounting for approximately 17 million deaths per year. Despite conventional treatment, cardiac tissue engineering emerges as a potential alternative for the advancement and treatment of these patients, using biomaterials to replace or repair cardiac tissues. Among these materials, gelatin in its methacrylated form (GelMA) is a biodegradable and biocompatible polymer with adjustable biophysical properties. Furthermore, gelatin has the ability to replace and perform collagen-like functions for cell development in vitro. The interest in using GelMA hydrogels combined with nanomaterials is increasingly growing to promote the responsiveness to external stimuli and improve certain properties of these hydrogels by exploring the incorporation of nanomaterials into these hydrogels to serve as electrical signaling conductive elements. This review highlights the applications of electrically conductive nanomaterials associated with GelMA hydrogels for the development of structures for cardiac tissue engineering, by focusing on studies that report the combination of GelMA with nanomaterials, such as gold and carbon derivatives (carbon nanotubes and graphene), in addition to the possibility of applying these materials in 3D tissue engineering, developing new possibilities for cardiac studies.
Collapse
Affiliation(s)
- Erika S Lisboa
- University of Tiradentes (Unit) and Institute of Technology and Research (ITP), Av. Murilo Dantas, 300, 49010-390 Aracaju, Brazil
| | - Carine Serafim
- University of Tiradentes (Unit) and Institute of Technology and Research (ITP), Av. Murilo Dantas, 300, 49010-390 Aracaju, Brazil
| | - Wanessa Santana
- University of Tiradentes (Unit) and Institute of Technology and Research (ITP), Av. Murilo Dantas, 300, 49010-390 Aracaju, Brazil
| | - Victoria L S Dos Santos
- University of Tiradentes (Unit) and Institute of Technology and Research (ITP), Av. Murilo Dantas, 300, 49010-390 Aracaju, Brazil
| | - Ricardo L C de Albuquerque-Junior
- Post-Graduate Program in Dentistry, Department of Dentistry, Federal University of Santa Catarina, Florianópolis 88040-370, Brazil; Department of Pathology, Health Sciences Center, Federal University of Santa Catarina, Florianópolis 88040-370, Brazil
| | - Marco V Chaud
- Laboratory of Biomaterials and Nanotechnology of UNISO (LaBNUS), University of Sorocaba, Sorocaba, São Paulo, Brazil
| | - Juliana C Cardoso
- University of Tiradentes (Unit) and Institute of Technology and Research (ITP), Av. Murilo Dantas, 300, 49010-390 Aracaju, Brazil
| | - Sona Jain
- University of Tiradentes (Unit) and Institute of Technology and Research (ITP), Av. Murilo Dantas, 300, 49010-390 Aracaju, Brazil
| | - Patrícia Severino
- University of Tiradentes (Unit) and Institute of Technology and Research (ITP), Av. Murilo Dantas, 300, 49010-390 Aracaju, Brazil.
| | - Eliana B Souto
- Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, MEDTECH, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| |
Collapse
|
39
|
Zhu D, Peng T, Zhang Z, Guo S, Su Y, Zhang K, Wang J, Liu C. Mesenchymal stem cells overexpressing XIST induce macrophage M2 polarization and improve neural stem cell homeostatic microenvironment, alleviating spinal cord injury. J Tissue Eng 2024; 15:20417314231219280. [PMID: 38223166 PMCID: PMC10785713 DOI: 10.1177/20417314231219280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 11/22/2023] [Indexed: 01/16/2024] Open
Abstract
Spinal cord injury (SCI) is a significant cause of disability worldwide, with limited treatment options. This study investigated the potential of bone marrow-derived mesenchymal stem cells (BMSCs) modified with XIST lentiviral vector to modulate macrophage polarization and affect neural stem cell (NSC) microenvironment reconstruction following SCI. Bioinformatics analysis revealed that MID1 might be crucial for BMSCs' treatment of SCI. XIST overexpression enriched Zmynd8 to the promoter region of MID1 and inhibited MID1 transcription, which promoted macrophage M2 polarization. In vitro experiments showed that BMSCs-XIST promoted NSC proliferation, migration, differentiation, and axonal growth by inducing macrophage M2 polarization, suppressing inflammation, and accelerating the re-establishment of the homeostatic microenvironment of NSCs. In vivo, animal experiments confirmed that BMSCs-XIST significantly alleviated SCI by promoting NSC differentiation and axon formation in the injured area. The study demonstrated the potential of XIST-overexpressing BMSCs for treating SCI by regulating macrophage polarization and homeostasis of the NSC microenvironment. These findings provide new insights into the development of stem cell-based therapies for SCI.
Collapse
Affiliation(s)
- Dan Zhu
- Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, Xianning, P.R. China
| | - Tie Peng
- Xianning Medical College, Hubei University of Science and Technology, Xianning, P.R. China
| | - Zhenwang Zhang
- Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, Xianning, P.R. China
| | - Shuang Guo
- Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, Xianning, P.R. China
| | - Ying Su
- Xianning Medical College, Hubei University of Science and Technology, Xianning, P.R. China
| | - Kangwei Zhang
- Xianning Medical College, Hubei University of Science and Technology, Xianning, P.R. China
| | - Jiawei Wang
- Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, Xianning, P.R. China
| | - Chao Liu
- Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, Xianning, P.R. China
| |
Collapse
|
40
|
Sarma S, Deka DJ, Rajak P, Laloo D, Das T, Chetia P, Saha D, Bharali A, Deka B. Potential injectable hydrogels as biomaterials for central nervous system injury: A narrative review. IBRAIN 2023; 9:402-420. [PMID: 38680508 PMCID: PMC11045191 DOI: 10.1002/ibra.12137] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 10/20/2023] [Accepted: 10/27/2023] [Indexed: 05/01/2024]
Abstract
Numerous modalities exist through which the central nervous system (CNS) may sustain injury or impairment, encompassing traumatic incidents, stroke occurrences, and neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease. Presently available pharmacological and therapeutic interventions are incapable of restoring or regenerating damaged CNS tissue, leading to substantial unmet clinical needs among patients with CNS ailments or injuries. To address and facilitate the recovery of the impaired CNS, cell-based repair strategies encompass multiple mechanisms, such as neuronal replacement, therapeutic factor secretion, and the promotion of host brain plasticity. Despite the progression of cell-based CNS reparation as a therapeutic strategy throughout the years, substantial barriers have impeded its widespread implementation in clinical settings. The integration of cell technologies with advancements in regenerative medicine utilizing biomaterials and tissue engineering has recently facilitated the surmounting of several of these impediments. This comprehensive review presents an overview of distinct CNS conditions necessitating cell reparation, in addition to exploring potential biomaterial methodologies that enhance the efficacy of treating brain injuries.
Collapse
Affiliation(s)
- Santa Sarma
- Girijananda Chowdhury Institute of Pharmaceutical ScienceAssam Science and Technology UniversityGuwahatiAssamIndia
| | - Dhruva J. Deka
- Girijananda Chowdhury Institute of Pharmaceutical ScienceAssam Science and Technology UniversityGuwahatiAssamIndia
| | - Prakash Rajak
- Department of Pharmaceutical SciencesDibrugarh UniversityDibrugarhAssamIndia
| | - Damiki Laloo
- School of Pharmaceutical SciencesGirijananda Chowdhury UniversityGuwahatiAssamIndia
| | - Trishna Das
- School of Pharmaceutical SciencesGirijananda Chowdhury UniversityGuwahatiAssamIndia
| | - Purbajit Chetia
- Department of PharmacologyNETES Institute of Pharmaceutical Science, Nemcare Group of Institutes, MirzaGuwahatiAssamIndia
| | - Dipankar Saha
- School of Pharmaceutical SciencesGirijananda Chowdhury UniversityGuwahatiAssamIndia
| | - Alakesh Bharali
- Department of Pharmaceutical SciencesDibrugarh UniversityDibrugarhAssamIndia
- School of Pharmaceutical SciencesGirijananda Chowdhury UniversityGuwahatiAssamIndia
| | - Bhargab Deka
- School of Pharmaceutical SciencesGirijananda Chowdhury UniversityGuwahatiAssamIndia
| |
Collapse
|
41
|
Yoon J, Han H, Jang J. Nanomaterials-incorporated hydrogels for 3D bioprinting technology. NANO CONVERGENCE 2023; 10:52. [PMID: 37968379 PMCID: PMC10651626 DOI: 10.1186/s40580-023-00402-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 10/24/2023] [Indexed: 11/17/2023]
Abstract
In the field of tissue engineering and regenerative medicine, various hydrogels derived from the extracellular matrix have been utilized for creating engineered tissues and implantable scaffolds. While these hydrogels hold immense promise in the healthcare landscape, conventional bioinks based on ECM hydrogels face several challenges, particularly in terms of lacking the necessary mechanical properties required for 3D bioprinting process. To address these limitations, researchers are actively exploring novel nanomaterial-reinforced ECM hydrogels for both mechanical and functional aspects. In this review, we focused on discussing recent advancements in the fabrication of engineered tissues and monitoring systems using nanobioinks and nanomaterials via 3D bioprinting technology. We highlighted the synergistic benefits of combining numerous nanomaterials into ECM hydrogels and imposing geometrical effects by 3D bioprinting technology. Furthermore, we also elaborated on critical issues remaining at the moment, such as the inhomogeneous dispersion of nanomaterials and consequent technical and practical issues, in the fabrication of complex 3D structures with nanobioinks and nanomaterials. Finally, we elaborated on plausible outlooks for facilitating the use of nanomaterials in biofabrication and advancing the function of engineered tissues.
Collapse
Affiliation(s)
- Jungbin Yoon
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, South Korea
| | - Hohyeon Han
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, South Korea
| | - Jinah Jang
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, South Korea.
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, South Korea.
- Department of Convergence IT Engineering, Pohang University of Science and Technology (POSTECH), Pohang, South Korea.
- Institute of Convergence Science, Yonsei University, Seoul, South Korea.
| |
Collapse
|
42
|
Liu C, Liu W, Qi B, Fan L, Liu S, Yang Q, Yang Y, Yang S, Zhang Y, Wei X, Zhu L. Bone Homeostasis Modulating Orthopedic Adhesive for the Closed-Loop Management of Osteoporotic Fractures. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2302704. [PMID: 37605327 DOI: 10.1002/smll.202302704] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/26/2023] [Indexed: 08/23/2023]
Abstract
Patients with osteoporotic fractures often require effective fixation and subsequent bone repair. However, currently available materials are often limited functionally, failing to improve this cohort's outcomes. Herein, kaempferol-loaded mesoporous bioactive glass nanoparticles (MBGNs)-doped orthopedic adhesives are prepared to assist osteoporotic fracture fixation and restore dysregulated bone homeostasis, including promoting osteoblast formation while inhibiting osteoclastic bone-resorbing activity to synergistically promote osteoporotic fracture healing. The injectability, reversible adhesiveness and malleable properties endowed the orthopedic adhesives with high flexibility and hemostatic performance to adapt to complex clinical scenarios. Moreover, Ca2+ and SiO4 4- ions released from MBGNs can accelerate osteogenesis via the PI3K/AKT pathway, while kaempferol mediated osteoclastogenesis inhibition and can slow down the bone resorption process through NF-κB pathway, which regulated bone regeneration and remodeling. Importantly, implementing the orthopedic adhesive is validated as an effective closed-loop management approach in restoring the dysregulated bone homeostasis of osteoporotic fractures.
Collapse
Affiliation(s)
- Can Liu
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, 100102, China
- Department of Orthopedics, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510515, China
| | - Weilu Liu
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Baoyu Qi
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, 100102, China
| | - Lei Fan
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Shencai Liu
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Qinfeng Yang
- Division of Orthopaedic Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yusheng Yang
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Shuofei Yang
- Department of Vascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China
| | - Yili Zhang
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xu Wei
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, 100102, China
| | - Liguo Zhu
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, 100102, China
| |
Collapse
|
43
|
Li J, Zhang J, Gao Y, Lei S, Wu J, Chen X, Wang K, Duan X, Men K. Targeted siRNA Delivery by Bioinspired Cancer Cell Membrane-Coated Nanoparticles with Enhanced Anti-Cancer Immunity. Int J Nanomedicine 2023; 18:5961-5982. [PMID: 37901359 PMCID: PMC10612485 DOI: 10.2147/ijn.s429036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 10/07/2023] [Indexed: 10/31/2023] Open
Abstract
Introduction Cell-membrane nanocarriers are usually constructed by modifying the nanoparticle surface with cell membrane extracts, which has a direct benefit in endowing targeting capacity to nanocarriers based on their original cell types. However, delivering nucleic acid cargos by cell membrane-based nanoparticles is difficult owing to the strong negative charge of the cell membrane fraction. In this study, we developed a cancer cell membrane-based drug delivery system, the cMDS, for efficient siRNA delivery. Meanwhile, the cancer-specific immune response stimulated by the gene vector itself could offer synergistic anti-cancer ability. Methods The cMDS was prepared by ultrasound, and its transfection efficiency and anti-cancer ability were examined using cultures of CT26 cells. MTT and red blood cell hemolysis tests were performed to assess the safety of cMDS, while its targeted gene delivery and strong immune stimulation were investigated in a subcutaneous tumor model. Moreover, the detailed anti-cancer immune stimulation mechanisms of cMDS are uncovered by protein chip analysis. Results The cMDS was spherical core-shell structure. It showed high transfection efficiency and anti-cancer ability in vitro. In animal experiments, intravenously administered cMDS/siStat3 complex efficiently suppress the growth of colon cancer. Moreover, the result of protein chip analysis suggested that cMDS affect the migration and chemotaxis of immune cells. Conclusion The cMDS shows obvious tumor tissue-specific accumulation properties and strong immune stimulation ability. It is an advanced targeted gene delivery system with potent immunotherapeutic properties.
Collapse
Affiliation(s)
- Jingmei Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Jin Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Yan Gao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Sibei Lei
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Jieping Wu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Xiaohua Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Kaiyu Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Xingmei Duan
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, People’s Republic of China
| | - Ke Men
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| |
Collapse
|
44
|
Walsh CM, Wychowaniec JK, Costello L, Brougham DF, Dooley D. An In Vitro and Ex Vivo Analysis of the Potential of GelMA Hydrogels as a Therapeutic Platform for Preclinical Spinal Cord Injury. Adv Healthc Mater 2023; 12:e2300951. [PMID: 37114899 PMCID: PMC11468190 DOI: 10.1002/adhm.202300951] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Indexed: 04/29/2023]
Abstract
Spinal cord injury (SCI) is a devastating condition with no curative therapy currently available. Immunomodulation can be applied as a therapeutic strategy to drive alternative immune cell activation and promote a proregenerative injury microenvironment. Locally injected hydrogels carrying immunotherapeutic cargo directly to injured tissue offer an encouraging treatment approach from an immunopharmacological perspective. Gelatin methacrylate (GelMA) hydrogels are promising in this regard, however, detailed analysis on the immunogenicity of GelMA in the specific context of the SCI microenvironment is lacking. Here, the immunogenicity of GelMA hydrogels formulated with a translationally relevant photoinitiator is analyzed in vitro and ex vivo. 3% (w/v) GelMA, synthesized from gelatin type-A, is first identified as the optimal hydrogel formulation based on mechanical properties and cytocompatibility. Additionally, 3% GelMA-A does not alter the expression profile of key polarization markers in BV2 microglia or RAW264.7 macrophages after 48 h. Finally, it is shown for the first time that 3% GelMA-A can support the ex vivo culture of primary murine organotypic spinal cord slices for 14 days with no direct effect on glial fibrillary acidic protein (GFAP+ ) astrocyte or ionized calcium-binding adaptor molecule 1 (Iba-1+ ) microglia reactivity. This provides evidence that GelMA hydrogels can act as an immunotherapeutic hydrogel-based platform for preclinical SCI.
Collapse
Affiliation(s)
- Ciara M. Walsh
- School of MedicineHealth Sciences CentreUniversity College DublinBelfieldDublinD04 V1W8Ireland
- UCD Conway Institute of Biomolecular & Biomedical ResearchUniversity College DublinBelfieldDublinD04 V1W8Ireland
| | - Jacek K. Wychowaniec
- School of ChemistryUniversity College DublinBelfieldDublinD04 V1W8Ireland
- AO Research Institute DavosClavadelerstrasse 8Davos7270Switzerland
| | - Louise Costello
- School of MedicineHealth Sciences CentreUniversity College DublinBelfieldDublinD04 V1W8Ireland
| | - Dermot F. Brougham
- School of ChemistryUniversity College DublinBelfieldDublinD04 V1W8Ireland
| | - Dearbhaile Dooley
- School of MedicineHealth Sciences CentreUniversity College DublinBelfieldDublinD04 V1W8Ireland
- UCD Conway Institute of Biomolecular & Biomedical ResearchUniversity College DublinBelfieldDublinD04 V1W8Ireland
| |
Collapse
|
45
|
Kwokdinata C, Ramanujam V, Chen J, de Oliveira PN, Nai MH, Chooi WH, Lim CT, Ng SY, David L, Chew SY. Encapsulation of Human Spinal Cord Progenitor Cells in Hyaluronan-Gelatin Hydrogel for Spinal Cord Injury Treatment. ACS APPLIED MATERIALS & INTERFACES 2023; 15:50679-50692. [PMID: 37751213 DOI: 10.1021/acsami.3c07419] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/27/2023]
Abstract
Transplanting human induced pluripotent stem cells (iPSCs)-derived spinal cord progenitor cells (SCPCs) is a promising approach to treat spinal cord injuries. However, stem cell therapies face challenges in cell survival, cell localization to the targeted site, and the control of cell differentiation. Here, we encapsulated SCPCs in thiol-modified hyaluronan-gelatin hydrogels and optimized scaffold mechanical properties and cell encapsulation density to promote cell viability and neuronal differentiation in vitro and in vivo. Different compositions of hyaluronan-gelatin hydrogels formulated by varying concentrations of poly(ethylene glycol) diacrylate were mechanically characterized by using atomic force microscopy. In vitro SCPC encapsulation study showed higher cell viability and proliferation with lower substrate Young's modulus (200 Pa vs 580 Pa) and cell density. Moreover, the soft hydrogels facilitated a higher degree of neuronal differentiation with extended filament structures in contrast to clumped cellular morphologies obtained in stiff hydrogels (p < 0.01). When transplanted in vivo, the optimized SCPC-encapsulated hydrogels resulted in higher cell survival and localization at the transplanted region as compared to cell delivery without hydrogel encapsulation at 2 weeks postimplantation within the rat spinal cord (p < 0.01). Notably, immunostaining demonstrated that the hydrogel-encapsulated SCPCs differentiated along the neuronal and oligodendroglial lineages in vivo. The lack of pluripotency and proliferation also supported the safety of the SCPC transplantation approach. Overall, the injectable hyaluronan-gelatin hydrogel shows promise in supporting the survival and neural differentiation of human SCPCs after transplantation into the spinal cord.
Collapse
Affiliation(s)
- Christy Kwokdinata
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637459, Singapore
| | - Vaibavi Ramanujam
- CNRS@CREATE, Create Tower #08-01, 1 Create Way, Singapore 138602, Singapore
| | - Jiahui Chen
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637459, Singapore
| | | | - Mui Hoon Nai
- Department of Biomedical Engineering, National University of Singapore, Singapore 117576, Singapore
| | - Wai Hon Chooi
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Republic of Singapore
| | - Chwee Teck Lim
- Department of Biomedical Engineering, National University of Singapore, Singapore 117576, Singapore
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
| | - Shi Yan Ng
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Republic of Singapore
| | - Laurent David
- CNRS@CREATE, Create Tower #08-01, 1 Create Way, Singapore 138602, Singapore
- Ingénierie des Matériaux Polymères IMP UMR 5223, CNRS, Université Claude Bernard Lyon 1, INSA de Lyon, Université Jean Monnet, Université de Lyon, Villeurbanne F69622, France
| | - Sing Yian Chew
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637459, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| |
Collapse
|
46
|
Fang Q, Wei Y, Zhang Y, Cao W, Yan L, Kong M, Zhu Y, Xu Y, Guo L, Zhang L, Wang W, Yu Y, Sun J, Yang J. Stem cells as potential therapeutics for hearing loss. Front Neurosci 2023; 17:1259889. [PMID: 37746148 PMCID: PMC10512725 DOI: 10.3389/fnins.2023.1259889] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 08/23/2023] [Indexed: 09/26/2023] Open
Abstract
Hearing impairment is a global health problem. Stem cell therapy has become a cutting-edge approach to tissue regeneration. In this review, the recent advances in stem cell therapy for hearing loss have been discussed. Nanomaterials can modulate the stem cell microenvironment to augment the therapeutic effects further. The potential of combining nanomaterials with stem cells for repairing and regenerating damaged inner ear hair cells (HCs) and spiral ganglion neurons (SGNs) has also been discussed. Stem cell-derived exosomes can contribute to the repair and regeneration of damaged tissue, and the research progress on exosome-based hearing loss treatment has been summarized as well. Despite stem cell therapy's technical and practical limitations, the findings reported so far are promising and warrant further investigation for eventual clinical translation.
Collapse
Affiliation(s)
- Qiaojun Fang
- Department of Otolaryngology-Head and Neck Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- School of Life Sciences and Technology, Southeast University, Nanjing, China
| | - Yongjie Wei
- Department of Otolaryngology-Head and Neck Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yuhua Zhang
- Department of Otolaryngology-Head and Neck Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Wei Cao
- Department of Otolaryngology-Head and Neck Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Lin Yan
- Department of Otolaryngology-Head and Neck Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Mengdie Kong
- School of Life Sciences and Technology, Southeast University, Nanjing, China
| | - Yongjun Zhu
- Department of Otolaryngology-Head and Neck Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yan Xu
- Department of Otolaryngology-Head and Neck Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Lingna Guo
- Department of Otolaryngology-Head and Neck Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Lei Zhang
- Department of Otolaryngology-Head and Neck Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Weiqing Wang
- Department of Otolaryngology-Head and Neck Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yafeng Yu
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jingwu Sun
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Jianming Yang
- Department of Otolaryngology-Head and Neck Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
47
|
Jia Z, Zeng H, Ye X, Dai M, Tang C, Liu L. Hydrogel-based treatments for spinal cord injuries. Heliyon 2023; 9:e19933. [PMID: 37809859 PMCID: PMC10559361 DOI: 10.1016/j.heliyon.2023.e19933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/05/2023] [Accepted: 09/06/2023] [Indexed: 10/10/2023] Open
Abstract
Spinal cord injury (SCI) is characterized by damage resulting in dysfunction of the spinal cord. Hydrogels are common biomaterials that play an important role in the treatment of SCI. Hydrogels are biocompatible, and some have electrical conductivity that are compatible with spinal cord tissues. Hydrogels have a high drug-carrying capacity, allowing them to be used for SCI treatment through the loading of various types of active substances, drugs, or cells. We first discuss the basic anatomy and physiology of the human spinal cord and briefly discuss SCI and its treatment. Then, we describe different treatment strategies for SCI. We further discuss the crosslinking methods and classification of hydrogels and detail hydrogel biomaterials prepared using different processing methods for the treatment of SCI. Finally, we analyze the future applications and limitations of hydrogels for SCI. The development of biomaterials opens up new possibilities and options for the treatment of SCI. Thus, our findings will inspire scholars in related fields and promote the development of hydrogel therapy for SCI.
Collapse
Affiliation(s)
- Zhiqiang Jia
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, China
| | - Huanxuan Zeng
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, China
| | - Xiuzhi Ye
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, China
| | - Minghai Dai
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, China
| | - Chengxuan Tang
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, China
| | - Liangle Liu
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, China
| |
Collapse
|
48
|
He J, Sun Y, Gao Q, He C, Yao K, Wang T, Xie M, Yu K, Nie J, Chen Y, He Y. Gelatin Methacryloyl Hydrogel, from Standardization, Performance, to Biomedical Application. Adv Healthc Mater 2023; 12:e2300395. [PMID: 37115708 DOI: 10.1002/adhm.202300395] [Citation(s) in RCA: 79] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/23/2023] [Indexed: 04/29/2023]
Abstract
Gelatin methacryloyl (GelMA), a photocurable hydrogel, is widely used in 3D culture, particularly in 3D bioprinting, due to its high biocompatibility, tunable physicochemical properties, and excellent formability. However, as the properties and performances of GelMA vary under different synthetic conditions, there is a lack of standardization, leading to conflicting results. In this study, a uniform standard is established to understand and enhance GelMA applications. First, the basic concept of GelMA and the density of the molecular network (DMN) are defined. Second, two properties, degrees of substitution and ratio of solid content, as the main measurable parameters determining the DMN are used. Third, the mechanisms and relationships between DMN and its performance in various applications in terms of porosity, viscosity, formability, mechanical strength, swelling, biodegradation, and cytocompatibility are theoretically explained. The main questions that are answered: what does performance mean, why is it important, how to optimize the basic parameters to improve the performance, and how to characterize it reasonably and accurately? Finally, it is hoped that this knowledge will eliminate the need for researchers to conduct tedious and repetitive pre-experiments, enable easy communication for achievements between groups under the same standard, and fully explore the potential of the GelMA hydrogel.
Collapse
Affiliation(s)
- Jing He
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yuan Sun
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Qing Gao
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
- Engineering for Life Group (EFL), Suzhou, 215101, China
| | - Chanfan He
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Ke Yao
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Tongyao Wang
- State Key Laboratory of Catalysis, National Laboratory for Clean Energy, 2011-Collaborative Innovation Center of Chemistry for Energy Materials, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Mingjun Xie
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
- Plastic and Reconstructive Surgery Center, Department of Plastic and Reconstructive Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Kang Yu
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Jing Nie
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yuewei Chen
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yong He
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
- Engineering for Life Group (EFL), Suzhou, 215101, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, College of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| |
Collapse
|
49
|
Wang H, Tang Q, Lu Y, Chen C, Zhao YL, Xu T, Yang CW, Chen XQ. Berberine-loaded MSC-derived sEVs encapsulated in injectable GelMA hydrogel for spinal cord injury repair. Int J Pharm 2023; 643:123283. [PMID: 37536642 DOI: 10.1016/j.ijpharm.2023.123283] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/08/2023] [Accepted: 07/28/2023] [Indexed: 08/05/2023]
Abstract
After spinal cord injury (SCI), local inflammatory response and fibrous scar formation severely hinder nerve regeneration. Berberine (Ber) has a powerful regulatory effect on the local microenvironment, but its limited solubility and permeability through the blood-brain barrier severely limit its systemic efficacy. Human umbilical cord mesenchymal stem cells (hUC-MSCs)-derived small extracellular vesicles (sEVs) are natural nanocarriers with high cargo loading capacity, and can cross the blood-brain barrier. Most importantly, sEVs can improve drug solubility and drug utilization. Therefore, they can overcome many defects of Ber application. This experiment aimed to design a Ber-carrying hUC-MSCs-derived sEVs and GelMA hydrogel. Ber was loaded into sEVs (sEVs-Ber) by ultrasonic co-incubation with a drug loading capacity (LC) of 15.07%. The unhindered release of up to 80% of sEVs-Ber from GelMA hydrogel was accomplished for up to 14 days. And they could be directly absorbed by local cells of injury, allowing for direct local delivery of the drug and enhancing its efficacy. The experimental results confirmed injecting GelMA-sEVs-Ber into spinal cord defects could exert anti-inflammatory effects by regulating the expression of inflammatory factors. It also demonstrated the anti-fibrotic effect of Ber in SCI for the first time. The modulatory effects of sEVs and Ber on the local microenvironment significantly promoted nerve regeneration and recovery of motor function in post-SCI rats. These results demonstrated that the GelMA-sEVs-Ber dual carrier system is a promising therapeutic strategy for SCI repair.
Collapse
Affiliation(s)
- Heng Wang
- Department of Orthopedics, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China.
| | - Qin Tang
- Department of Orthopedics, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Yang Lu
- Department of Orthopedics, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Cheng Chen
- Department of Orthopedics, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Yu-Lin Zhao
- Department of Orthopedics, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Tao Xu
- Department of Orthopedics, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China.
| | - Chang-Wei Yang
- Department of Orthopedics, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China.
| | - Xiao-Qing Chen
- Department of Orthopedics, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China.
| |
Collapse
|
50
|
Wan J, He Z, Peng R, Wu X, Zhu Z, Cui J, Hao X, Chen A, Zhang J, Cheng P. Injectable photocrosslinking spherical hydrogel-encapsulated targeting peptide-modified engineered exosomes for osteoarthritis therapy. J Nanobiotechnology 2023; 21:284. [PMID: 37605203 PMCID: PMC10440922 DOI: 10.1186/s12951-023-02050-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 08/04/2023] [Indexed: 08/23/2023] Open
Abstract
Osteoarthritis (OA) is a common degenerative joint disease urgently needing effective treatments. Bone marrow mesenchymal stromal cell-derived exosomes (Exo) are considered good drug carriers whereas they have limitations such as fast clearance and low retention. This study aimed to overcome the limitations of Exo in drug delivery using multiple strategies. Novel photocrosslinking spherical gelatin methacryloyl hydrogel (GelMA)-encapsulated cartilage affinity WYRGRL (W) peptide-modified engineered Exo were developed for OA treatment and the performance of the engineered Exo (W-Exo@GelMA) loaded with a small inhibitor LRRK2-IN-1 (W-Exo-L@GelMA) was investigated in vitro and in vivo. The W-Exo-L@GelMA showed an effective targeting effect on chondrocytes and a pronounced action on suppressing catabolism and promoting anabolism in vitro. Moreover, W-Exo-L@GelMA remarkably inhibited OA-related inflammation and immune gene expression, rescuing the IL-1β-induced transcriptomic responses. With enhanced retention in the joint, W-Exo-L@GelMA demonstrated superior anti-OA activity and cartilage repair ability in the OA murine model. The therapeutic effect was validated in the cultured human OA cartilage. In conclusion, photocrosslinking spherical hydrogel-encapsulated targeting peptide-modified engineered Exo exhibit notable potential in OA therapy. Engineering Exo by a series of strategies enhanced the targeting ability and retention and cartilage-targeting and Exo-mediated drug delivery may offer a novel strategy for OA treatment.Clinical trial registration: Not applciable.
Collapse
Affiliation(s)
- Junlai Wan
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095#, Jie-Fang Avenue, Qiaokou District, Wuhan, 430030, Hubei, China
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Zhiyi He
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095#, Jie-Fang Avenue, Qiaokou District, Wuhan, 430030, Hubei, China
| | - Renpeng Peng
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095#, Jie-Fang Avenue, Qiaokou District, Wuhan, 430030, Hubei, China
| | - Xiaopei Wu
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, 430070, China
| | - Ziqing Zhu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095#, Jie-Fang Avenue, Qiaokou District, Wuhan, 430030, Hubei, China
| | - Jiarui Cui
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Xiaoxia Hao
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Anmin Chen
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095#, Jie-Fang Avenue, Qiaokou District, Wuhan, 430030, Hubei, China.
| | - Jiaming Zhang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095#, Jie-Fang Avenue, Qiaokou District, Wuhan, 430030, Hubei, China.
| | - Peng Cheng
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095#, Jie-Fang Avenue, Qiaokou District, Wuhan, 430030, Hubei, China.
| |
Collapse
|