1
|
Luo Z, Shangguan Z, Cao L, Zhang Y, Li Q, Shi X, Fu J, Wang C, Dou X, Tan W, Li Q. Cerebrospinal fluid-contacting neurons: a promising source for adult neural stem cell transplantation in spinal cord injury treatment. Front Cell Dev Biol 2025; 13:1549194. [PMID: 40143967 PMCID: PMC11936957 DOI: 10.3389/fcell.2025.1549194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 02/14/2025] [Indexed: 03/28/2025] Open
Abstract
Transplantation of adult neural stem cells (NSCs) is regarded as one of the most promising approaches for treating spinal cord injury (SCI). However, securing a sufficient and reliable source of adult NSCs remains one of the primary challenges in applying this method for SCI treatment. Cerebrospinal fluid-contacting neurons (CSF-cNs) act as adult NSCs and can be substantially expanded in vitro while maintaining their NSC characteristics even after 60 passages. When CSF-cNs are transplanted into the injury sites of SCI mice, they demonstrate high survival rates along with the ability to proliferate and differentiate into neurons, astrocytes, and oligodendrocytes. Additionally, significant improvements in motor function have been observed in SCI mice following the transplantation of CSF-cNs. These results suggest that CSF-cNs may represent a promising source of adult NSCs for transplantation therapy in SCI.
Collapse
Affiliation(s)
- Zhangrong Luo
- Department of Emergency, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- Department of Traumatic Orthopedics, The Affiliated Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Zeyu Shangguan
- Department of Emergency, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Liang Cao
- Department of Emergency, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Yi Zhang
- Department of Emergency, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Qizhe Li
- Department of Emergency, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Xuexing Shi
- Department of Emergency, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Jiangquan Fu
- Department of Emergency, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Chunqing Wang
- Department of Emergency, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xiaowei Dou
- Clinical Research Center, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Wei Tan
- Department of Emergency, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Qing Li
- Department of Emergency, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
2
|
Yue X, Chen X, Zang Y, Wu J, Chen G, Tan H, Yang K. Bioinformatics analysis reveals key mechanisms of oligodendrocytes and oligodendrocyte precursor cells regulation in spinal cord Injury. Sci Rep 2025; 15:6400. [PMID: 39984610 PMCID: PMC11845783 DOI: 10.1038/s41598-025-90489-z] [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: 10/11/2024] [Accepted: 02/13/2025] [Indexed: 02/23/2025] Open
Abstract
Despite extensive research, spinal cord injuries (SCI), which could cause severe sensory, motor and autonomic dysfunction, remain largely incurable. Oligodendrocytes and oligodendrocyte precursor cells (ODC/OPC) play a crucial role in neural morphological repair and functional recovery following SCI. We performed single-cell sequencing (scRNA-seq) on 59,558 cells from 39 mouse samples, combined with microarray data from 164 SCI samples and 3 uninjured samples. We further validated our findings using a large clinical cohort consisting of 38 SCI patients, 10 healthy controls, and 10 trauma controls, assessed with the American Spinal Cord Injury Association (ASIA) scale. We proposed a novel SCI classification model based on the expression of prognostic differentially expressed ODC/OPC differentiation-related genes (PDEODGs). This model includes three types: Low ODC/OPC Score Classification (LOSC), Median ODC/OPC Score Classification (MOSC), and High ODC/OPC Score Classification (HOSC). Considering the relationship between these subtypes and prognosis, we speculated that enhancing ODC/OPC differentiation and inhibiting inflammatory infiltration may improve outcomes. Additionally, we identified potential treatments for SCI that target key genes within these subtypes, offering promising implications for therapy.
Collapse
Affiliation(s)
- Xi Yue
- Department of Orthopedics, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xunling Chen
- Children's Hospital Affiliated of Zhengzhou University, Zhengzhou, China
| | - Yang Zang
- Department of Orthopedics, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jinliang Wu
- Department of Orthopedics, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guanhao Chen
- Department of Orthopedics, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongyu Tan
- Department of Orthopedics, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Kerong Yang
- Department of Orthopedics, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
3
|
Liao YH, Tian MH, Zhou WY, He BQ, Tang C, Tang Q, Ye RP, Zhong DJ. Low-intensity pulsed ultrasound promotes proliferation and differentiation of neural stem cells to enhance spinal cord injury recovery. Mol Biol Rep 2025; 52:245. [PMID: 39964570 DOI: 10.1007/s11033-025-10333-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 02/03/2025] [Indexed: 05/10/2025]
Abstract
BACKGROUND Neural stem cells (NSCs) are known for their high capacity for proliferation, self-renewal, and multidirectional differentiation. However, they often fail to survive or differentiate into mature neurons capable of replacing lost neurons. This study evaluated the potential role of low-intensity pulsed ultrasound (LIPUS) in promoting NSC proliferation and differentiation both in vitro and in vivo, as well as its role in enhancing spinal cord injury (SCI) recovery. METHODS NSCs were isolated, stimulated with LIPUS, and characterized through identification and detection assays. The safety and efficacy of LIPUS in promoting NSC proliferation and differentiation were assessed through cell viability and apoptosis assays, and neuronal marker expression analysis. In vivo, NSCs encoding fluorescent proteins were transplanted into a rat model of SCI. The SCI rats received LIPUS for 4 weeks. Later, functional recovery, morphological changes and neuronal structures were evaluated. RESULT The isolated NSCs were successfully identified. LIPUS significantly enhanced NSC proliferation and increased the expression of key neurogenic markers and neurotrophic factors, while reducing GFAP expression and avoiding apoptosis. In vivo, the NSCs/LIPUS + group demonstrated higher survival and differentiation of transplanted NSCs, along with improved BBB scores and enhanced neural marker expression compared with the NSCs/LIPUS - group. CONCLUSION LIPUS stimulation effectively promoted NSC proliferation and differentiation and enhanced the survival and function of transplanted NSCs at the SCI site, leading to improved SCI recovery.
Collapse
Affiliation(s)
- Ye-Hui Liao
- Department of Orthopedics, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Min-Hao Tian
- Department of Orthopedics, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Wen-Yang Zhou
- Department of Orthopedics, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Bao-Qiang He
- Department of Orthopedics, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Chao Tang
- Department of Orthopedics, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Qiang Tang
- Department of Orthopedics, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Ru-Pei Ye
- Pathology Department, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, China.
| | - De-Jun Zhong
- Department of Orthopedics, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, China.
| |
Collapse
|
4
|
Alsiraey N, Dewald HD. Nitroxidative stress in human neural progenitor cells: In situ measurement of nitric oxide/peroxynitrite imbalance using metalloporphyrin nanosensors. J Inorg Biochem 2025; 263:112785. [PMID: 39603147 DOI: 10.1016/j.jinorgbio.2024.112785] [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: 10/10/2024] [Revised: 11/18/2024] [Accepted: 11/21/2024] [Indexed: 11/29/2024]
Abstract
Nitric oxide (NO) is an essential inorganic signaling molecule produced by constitutive NO synthase (cNOS) in the neurological system. Under pathological conditions, NO rapidly reacts with superoxide (O2•-) to generate peroxynitrite (ONOO¯). Elevated ONOO¯ concentrations induce nitroxidative stress, potentially contributing to numerous pathological processes as observed in neurodegenerative diseases including Alzheimer's disease (AD). Metalloporphyrin nanosensors, (200-300 nm diameter), were applied to quantify the NO/ONOO¯ balance produced by a single human neural progenitor cell (hNPC), in situ. These nanosensors, positioned in proximity of 4-5 ± 1 μm from the hNPCs membrane, enabled real-time measurement of NO and ONOO¯ concentrations following calcium ionophore (CaI) stimulation. The ratio of NO to ONOO¯ concentration ([NO]/[ONOO¯]) was established for the purpose of quantifying nitroxidative stress levels. Normal hNPCs produced a maximum of 107 ± 1 nmol/L of NO and 451 ± 7 nmol/L of ONOO¯, yielding a [NO]/[ONOO¯] ratio of 0.25 ± 0.005. In contrast, the model of the dysfunctional hNPCs, for long-term (48 h) amyloid-beta 42 (Aβ42) exposure significantly altered NO/ONOO¯ production. The NO level decreased to 14 ± 0.1 nmol/L, while ONOO¯ increased to 843 ± 0.8 nmol/L, resulting in a 94 % reduction of the [NO]/[ONOO¯] ratio to 0.016 ± 0.0001. The [NO]/[ONOO¯] ratio is determined by this work as a possible biomarker of nNOS efficiency and hNPC dysfunction, with implications for neurodegenerative disorders such as AD. Promising applications in the early medical diagnosis of neurological illnesses, electrochemical metalloporphyrin nanosensors demonstrate efficacy in real-time nitroxidative stress monitoring.
Collapse
Affiliation(s)
- Nouf Alsiraey
- Department of Chemistry and Biochemistry, Ohio University, Athens, OH 45701, USA; Department of Chemistry, College of Science, Northern Border University, Arar 91431, Saudi Arabia
| | - Howard D Dewald
- Department of Chemistry and Biochemistry, Ohio University, Athens, OH 45701, USA.
| |
Collapse
|
5
|
Shen YJ, Huang YC, Cheng YC. Advancements in Antioxidant-Based Therapeutics for Spinal Cord Injury: A Critical Review of Strategies and Combination Approaches. Antioxidants (Basel) 2024; 14:17. [PMID: 39857350 PMCID: PMC11763222 DOI: 10.3390/antiox14010017] [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: 12/01/2024] [Revised: 12/21/2024] [Accepted: 12/24/2024] [Indexed: 01/27/2025] Open
Abstract
Spinal cord injury (SCI) initiates a cascade of secondary damage driven by oxidative stress, characterized by the excessive production of reactive oxygen species and other reactive molecules, which exacerbate cellular and tissue damage through the activation of deleterious signaling pathways. This review provides a comprehensive and critical evaluation of recent advancements in antioxidant-based therapeutic strategies for SCI, including natural compounds, RNA-based therapies, stem cell interventions, and biomaterial applications. It emphasizes the limitations of single-regimen approaches, particularly their limited efficacy and suboptimal delivery to injured spinal cord tissue, while highlighting the synergistic potential of combination therapies that integrate multiple modalities to address the multifaceted pathophysiology of SCI. By analyzing emerging trends and current limitations, this review identifies key challenges and proposes future directions, including the refinement of antioxidant delivery systems, the development of multi-targeted approaches, and strategies to overcome the structural complexities of the spinal cord. This work underscores the pressing need for innovative and integrative therapeutic approaches to advance the clinical translation of antioxidant-based interventions and improve outcomes for SCI patients.
Collapse
Affiliation(s)
- Yang-Jin Shen
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Yin-Cheng Huang
- Neuroscience Research Center, Chang Gung Memorial Hospital, Linkou, Taoyuan 333423, Taiwan
- Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou Medical Center, Taoyuan 333423, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Yi-Chuan Cheng
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Neuroscience Research Center, Chang Gung Memorial Hospital, Linkou, Taoyuan 333423, Taiwan
| |
Collapse
|
6
|
Jeon J, Park SH, Choi J, Han SM, Kim HW, Shim SR, Hyun JK. Association between neural stem/progenitor cells and biomaterials in spinal cord injury therapies: A systematic review and network meta-analysis. Acta Biomater 2024; 183:50-60. [PMID: 38871200 DOI: 10.1016/j.actbio.2024.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 06/03/2024] [Accepted: 06/06/2024] [Indexed: 06/15/2024]
Abstract
Spinal cord injury (SCI) is associated with substantial healthcare challenges, frequently resulting in enduring sensory and motor deficits alongside various chronic complications. While advanced regenerative therapies have shown promise in preclinical research, their translation into clinical application has been limited. In response, this study utilized a comprehensive network meta-analysis to evaluate the effectiveness of neural stem/progenitor cell (NSPC) transplantation across animal models of SCI. We analyzed 363 outcomes from 55 distinct studies, categorizing the treatments into NSPCs alone (cell only), NSPCs with scaffolds (cell + scaffold), NSPCs with hydrogels (cell + hydrogel), standalone scaffolds (scaffold), standalone hydrogels (hydrogel), and control groups. Our analysis demonstrated significant enhancements in motor recovery, especially in gait function, within the NSPC treatment groups. Notably, the cell only group showed considerable improvements (standardized mean difference [SMD], 2.05; 95 % credible interval [CrI]: 1.08 to 3.10, p < 0.01), as did the cell + scaffold group (SMD, 3.73; 95 % CrI: 2.26 to 5.22, p < 0.001) and the cell + hydrogel group (SMD, 3.37; 95 % CrI: 1.02 to 5.78, p < 0.05) compared to controls. These therapeutic combinations not only reduced lesion cavity size but also enhanced neuronal regeneration, outperforming the cell only treatments. By integrating NSPCs with supportive biomaterials, our findings pave the way for refining these regenerative strategies to optimize their potential in clinical SCI treatment. Although there is no overall violation of consistency, the comparison of effect sizes between individual treatments should be interpreted in light of the inconsistency. STATEMENT OF SIGNIFICANCE: This study presents a comprehensive network meta-analysis exploring the efficacy of neural stem cell (NSC) transplantation, with and without biomaterials, in animal models of spinal cord injury (SCI). We demonstrate that NSCs, particularly when combined with biomaterials like scaffolds or hydrogels, significantly enhance motor and histological recovery post-SCI. These findings underscore the potential of NSC-based therapies, augmented with biomaterials, to advance SCI treatment, offering new insights into regenerative strategies that could significantly impact clinical practices.
Collapse
Affiliation(s)
- Jooik Jeon
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea; Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea
| | | | - Jonghyuk Choi
- Department of Preventive Medicine, College of Medicine, Dankook University, Cheonan 31116, Republic of Korea
| | - Sun Mi Han
- Medical record team, Konyang University Hospital, Daejeon 35365, Republic of Korea
| | - Hae-Won Kim
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea; Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea; Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan 31116, Republic of Korea
| | - Sung Ryul Shim
- Department of Biomedical Informatics, College of Medicine, Konyang University, Daejeon 35365, Republic of Korea.
| | - Jung Keun Hyun
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea; Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea; Wiregene, Co. Ltd., Osong 28160, Republic of Korea; Department of Rehabilitation Medicine, College of Medicine, Dankook University, Cheonan 31116, Republic of Korea.
| |
Collapse
|
7
|
Alsiraey N, Malinski T, Dewald HD. Using Metalloporphyrin Nanosensors for In Situ Monitoring and Measurement of Nitric Oxide and Peroxynitrite in a Single Human Neural Progenitor Cell. ACS Sens 2024; 9:3037-3047. [PMID: 38773722 DOI: 10.1021/acssensors.4c00234] [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: 05/24/2024]
Abstract
Nitric oxide (NO) is an inorganic signaling molecule that plays a crucial role in the regulation of numerous physiological functions. An oxidation product of the cytoprotective NO is cytotoxic peroxynitrite (ONOO-). In biological systems, the concentrations of NO and ONOO- are typically transient, ranging from nanomolar to micromolar, and these increases are normally followed by a swift return to their basal levels due to their short life spans. To understand the vital physiological role of NO and ONOO- in vitro and in vivo, sensitive and selective methods are necessary for direct and continuous NO and ONOO- measurements in real time. Because electrochemical methods can be adjusted for selectivity, sensitivity, and biocompatibility in demanding biological environments, they are suitable for real-time monitoring of NO and ONOO- release. Metalloporphyrin nanosensors, described here, have been designed to measure the concentration of NO and ONOO- produced by a single human neural progenitor cell (hNPC) in real time. These nanosensors (200-300 nm in diameter) can be positioned accurately in the proximity of 4-5 ± 1 μm from an hNPC membrane. The response time of the sensors is better than a millisecond, while detection limits for NO and ONOO- are 1 × 10-9 and 3 × 10-9 mol/L, respectively, with a linear concentration response of up to about 1 μM. The application of these metalloporphyrin nanosensors for the efficient measurement of the concentrations of NO and ONOO- in hNPCs is demonstrated, providing an opportunity to observe in real time the molecular changes of the two signaling molecules in situ.
Collapse
Affiliation(s)
- Nouf Alsiraey
- Department of Chemistry and Biochemistry, Ohio University, Athens, Ohio 45701, United States
- Department of Chemistry, Faculty of Science, Northern Border University, Arar 91431, Saudi Arabia
| | - Tadeusz Malinski
- Department of Chemistry and Biochemistry, Ohio University, Athens, Ohio 45701, United States
| | - Howard D Dewald
- Department of Chemistry and Biochemistry, Ohio University, Athens, Ohio 45701, United States
| |
Collapse
|
8
|
Chen YL, Feng XL, Tam KW, Fan CY, Cheung MPL, Yang YT, Wong S, Shum DKY, Chan YS, Cheung CW, Cheung M, Liu JA. Intrinsic and extrinsic actions of human neural progenitors with SUFU inhibition promote tissue repair and functional recovery from severe spinal cord injury. NPJ Regen Med 2024; 9:13. [PMID: 38519518 PMCID: PMC10959923 DOI: 10.1038/s41536-024-00352-4] [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: 07/09/2023] [Accepted: 02/06/2024] [Indexed: 03/25/2024] Open
Abstract
Neural progenitor cells (NPCs) derived from human pluripotent stem cells(hPSCs) provide major cell sources for repairing damaged neural circuitry and enabling axonal regeneration after spinal cord injury (SCI). However, the injury niche and inadequate intrinsic factors in the adult spinal cord restrict the therapeutic potential of transplanted NPCs. The Sonic Hedgehog protein (Shh) has crucial roles in neurodevelopment by promoting the formation of motorneurons and oligodendrocytes as well as its recently described neuroprotective features in response to the injury, indicating its essential role in neural homeostasis and tissue repair. In this study, we demonstrate that elevated SHH signaling in hNPCs by inhibiting its negative regulator, SUFU, enhanced cell survival and promoted robust neuronal differentiation with extensive axonal outgrowth, counteracting the harmful effects of the injured niche. Importantly, SUFU inhibition in NPCs exert non-cell autonomous effects on promoting survival and neurogenesis of endogenous cells and modulating the microenvironment by reducing suppressive barriers around lesion sites. The combined beneficial effects of SUFU inhibition in hNPCs resulted in the effective reconstruction of neuronal connectivity with the host and corticospinal regeneration, significantly improving neurobehavioral recovery in recipient animals. These results demonstrate that SUFU inhibition confers hNPCs with potent therapeutic potential to overcome extrinsic and intrinsic barriers in transplantation treatments for SCI.
Collapse
Affiliation(s)
- Yong-Long Chen
- Department of Anaesthesiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Xiang-Lan Feng
- Department of Anaesthesiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Kin-Wai Tam
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Chao-Yang Fan
- Department of Neuroscience, Tat Chee Avenue, City University of Hong Kong, Hong Kong, China
| | - May Pui-Lai Cheung
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yong-Ting Yang
- Department of Neuroscience, Tat Chee Avenue, City University of Hong Kong, Hong Kong, China
| | - Stanley Wong
- Department of Anaesthesiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Daisy Kwok-Yan Shum
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Ying-Shing Chan
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Chi-Wai Cheung
- Department of Anaesthesiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Hong Kong sanatorium hospital, Hong Kong, China
| | - Martin Cheung
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Jessica Aijia Liu
- Department of Anaesthesiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
- Department of Neuroscience, Tat Chee Avenue, City University of Hong Kong, Hong Kong, China.
| |
Collapse
|
9
|
Chudakova DA, Samoilova EM, Chekhonin VP, Baklaushev VP. Improving Efficiency of Direct Pro-Neural Reprogramming: Much-Needed Aid for Neuroregeneration in Spinal Cord Injury. Cells 2023; 12:2499. [PMID: 37887343 PMCID: PMC10605572 DOI: 10.3390/cells12202499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 10/13/2023] [Accepted: 10/18/2023] [Indexed: 10/28/2023] Open
Abstract
Spinal cord injury (SCI) is a medical condition affecting ~2.5-4 million people worldwide. The conventional therapy for SCI fails to restore the lost spinal cord functions; thus, novel therapies are needed. Recent breakthroughs in stem cell biology and cell reprogramming revolutionized the field. Of them, the use of neural progenitor cells (NPCs) directly reprogrammed from non-neuronal somatic cells without transitioning through a pluripotent state is a particularly attractive strategy. This allows to "scale up" NPCs in vitro and, via their transplantation to the lesion area, partially compensate for the limited regenerative plasticity of the adult spinal cord in humans. As recently demonstrated in non-human primates, implanted NPCs contribute to the functional improvement of the spinal cord after injury, and works in other animal models of SCI also confirm their therapeutic value. However, direct reprogramming still remains a challenge in many aspects; one of them is low efficiency, which prevents it from finding its place in clinics yet. In this review, we describe new insights that recent works brought to the field, such as novel targets (mitochondria, nucleoli, G-quadruplexes, and others), tools, and approaches (mechanotransduction and electrical stimulation) for direct pro-neural reprogramming, including potential ones yet to be tested.
Collapse
Affiliation(s)
- Daria A. Chudakova
- Federal Center for Brain and Neurotechnologies, Federal Medical and Biological Agency of Russia, 117513 Moscow, Russia
| | - Ekaterina M. Samoilova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Federal Research and Clinical Center of Specialised Medical Care and Medical Technologies FMBA of Russia, 115682 Moscow, Russia
| | - Vladimir P. Chekhonin
- Department of Medical Nanobiotechnology of Medical and Biological Faculty, Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, 117997 Moscow, Russia
| | - Vladimir P. Baklaushev
- Federal Center for Brain and Neurotechnologies, Federal Medical and Biological Agency of Russia, 117513 Moscow, Russia
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Federal Research and Clinical Center of Specialised Medical Care and Medical Technologies FMBA of Russia, 115682 Moscow, Russia
- Department of Medical Nanobiotechnology of Medical and Biological Faculty, Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, 117997 Moscow, Russia
| |
Collapse
|
10
|
Roh EJ, Kim DS, Kim JH, Lim CS, Choi H, Kwon SY, Park SY, Kim JY, Kim HM, Hwang DY, Han DK, Han I. Multimodal therapy strategy based on a bioactive hydrogel for repair of spinal cord injury. Biomaterials 2023; 299:122160. [PMID: 37209541 DOI: 10.1016/j.biomaterials.2023.122160] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 03/13/2023] [Accepted: 05/10/2023] [Indexed: 05/22/2023]
Abstract
Traumatic spinal cord injury results in permanent and serious neurological impairment, but there is no effective treatment yet. Tissue engineering approaches offer great potential for the treatment of SCI, but spinal cord complexity poses great challenges. In this study, the composite scaffold consists of a hyaluronic acid-based hydrogel, decellularized brain matrix (DBM), and bioactive compounds such as polydeoxyribonucleotide (PDRN), tumor necrosis factor-α/interferon-γ primed mesenchymal stem cell-derived extracellular vesicles (TI-EVs), and human embryonic stem cell-derived neural progenitor cells (NPC). The composite scaffold showed significant effects on regenerative prosses including angiogenesis, anti-inflammation, anti-apoptosis, and neural differentiation. In addition, the composite scaffold (DBM/PDRN/TI-EV/NPC@Gel) induced an effective spinal cord regeneration in a rat spinal cord transection model. Therefore, this multimodal approach using an integrated bioactive scaffold coupled with biochemical cues from PDRN and TI-EVs could be used as an advanced tissue engineering platform for spinal cord regeneration.
Collapse
Affiliation(s)
- Eun Ji Roh
- Department of Neurosurgery CHA University School of Medicine, 335 Pangyo-ro Bundang-gu, Seongnam-si, 13488, Republic of Korea; Department of Biomedical Science CHA University, 335 Pangyo-ro Bundang-gu, Seongnam-si, 13488, Republic of Korea
| | - Da-Seul Kim
- Department of Biomedical Science CHA University, 335 Pangyo-ro Bundang-gu, Seongnam-si, 13488, Republic of Korea; School of Integrative Engineering Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Jun Hyuk Kim
- Department of Biomedical Science CHA University, 335 Pangyo-ro Bundang-gu, Seongnam-si, 13488, Republic of Korea
| | - Chang Su Lim
- Department of Neurosurgery CHA University School of Medicine, 335 Pangyo-ro Bundang-gu, Seongnam-si, 13488, Republic of Korea
| | - Hyemin Choi
- Department of Neurosurgery CHA University School of Medicine, 335 Pangyo-ro Bundang-gu, Seongnam-si, 13488, Republic of Korea
| | - Su Yeon Kwon
- Department of Neurosurgery CHA University School of Medicine, 335 Pangyo-ro Bundang-gu, Seongnam-si, 13488, Republic of Korea
| | - So-Yeon Park
- Department of Biomedical Science CHA University, 335 Pangyo-ro Bundang-gu, Seongnam-si, 13488, Republic of Korea; Division of Biotechnology College of Life Sciences and Biotechnology Korea University, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Jun Yong Kim
- Department of Biomedical Science CHA University, 335 Pangyo-ro Bundang-gu, Seongnam-si, 13488, Republic of Korea
| | - Hyun-Mun Kim
- Department of Biomedical Science CHA University, 335 Pangyo-ro Bundang-gu, Seongnam-si, 13488, Republic of Korea
| | - Dong-Youn Hwang
- Department of Biomedical Science CHA University, 335 Pangyo-ro Bundang-gu, Seongnam-si, 13488, Republic of Korea
| | - Dong Keun Han
- Department of Biomedical Science CHA University, 335 Pangyo-ro Bundang-gu, Seongnam-si, 13488, Republic of Korea.
| | - Inbo Han
- Department of Neurosurgery CHA University School of Medicine, 335 Pangyo-ro Bundang-gu, Seongnam-si, 13488, Republic of Korea.
| |
Collapse
|
11
|
Wu Y, Tang Z, Zhang J, Wang Y, Liu S. Restoration of spinal cord injury: From endogenous repairing process to cellular therapy. Front Cell Neurosci 2022; 16:1077441. [PMID: 36523818 PMCID: PMC9744968 DOI: 10.3389/fncel.2022.1077441] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 11/08/2022] [Indexed: 09/26/2023] Open
Abstract
Spinal cord injury (SCI) disrupts neurological pathways and impacts sensory, motor, and autonomic nerve function. There is no effective treatment for SCI currently. Numerous endogenous cells, including astrocytes, macrophages/microglia, and oligodendrocyte, are involved in the histological healing process following SCI. By interfering with cells during the SCI repair process, some advancements in the therapy of SCI have been realized. Nevertheless, the endogenous cell types engaged in SCI repair and the current difficulties these cells confront in the therapy of SCI are poorly defined, and the mechanisms underlying them are little understood. In order to better understand SCI and create new therapeutic strategies and enhance the clinical translation of SCI repair, we have comprehensively listed the endogenous cells involved in SCI repair and summarized the six most common mechanisms involved in SCI repair, including limiting the inflammatory response, protecting the spared spinal cord, enhancing myelination, facilitating neovascularization, producing neurotrophic factors, and differentiating into neural/colloidal cell lines.
Collapse
Affiliation(s)
| | | | | | | | - Shengwen Liu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
12
|
Liu D, Fan B, Li J, Sun T, Ma J, Zhou X, Feng S. N6-methyladenosine modification: A potential regulatory mechanism in spinal cord injury. Front Cell Neurosci 2022; 16:989637. [PMID: 36212687 PMCID: PMC9539101 DOI: 10.3389/fncel.2022.989637] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 09/05/2022] [Indexed: 12/03/2022] Open
Abstract
N6-methyladenosine (m6A), an essential post-transcriptional modification in eukaryotes, is closely related to the development of pathological processes in neurological diseases. Notably, spinal cord injury (SCI) is a serious traumatic disease of the central nervous system, with a complex pathological mechanism which is still not completely understood. Recent studies have found that m6A modification levels are changed after SCI, and m6A-related regulators are involved in the changes of the local spinal cord microenvironment after injury. However, research on the role of m6A modification in SCI is still in the early stages. This review discusses the latest progress in the dynamic regulation of m6A modification, including methyltransferases (“writers”), demethylases (“erasers”) and m6A -binding proteins (“readers”). And then analyses the pathological mechanism relationship between m6A and the microenvironment after SCI. The biological processes involved included cell death, axon regeneration, and scar formation, which provides new insight for future research on the role of m6A modification in SCI and the clinical transformation of strategies for promoting recovery of spinal cord function.
Collapse
Affiliation(s)
- Derong Liu
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, Tianjin, China
| | - Baoyou Fan
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, Tianjin, China
| | - Jinze Li
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, Tianjin, China
| | - Tao Sun
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, Tianjin, China
| | - Jun Ma
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, Tianjin, China
| | - Xianhu Zhou
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
- *Correspondence: Xianhu Zhou,
| | - Shiqing Feng
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, Tianjin, China
- Shiqing Feng,
| |
Collapse
|
13
|
Guo W, Zhang X, Zhai J, Xue J. The roles and applications of neural stem cells in spinal cord injury repair. Front Bioeng Biotechnol 2022; 10:966866. [PMID: 36105599 PMCID: PMC9465243 DOI: 10.3389/fbioe.2022.966866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 07/28/2022] [Indexed: 12/05/2022] Open
Abstract
Spinal cord injury (SCI), which has no current cure, places a severe burden on patients. Stem cell-based therapies are considered promising in attempts to repair injured spinal cords; such options include neural stem cells (NSCs). NSCs are multipotent stem cells that differentiate into neuronal and neuroglial lineages. This feature makes NSCs suitable candidates for regenerating injured spinal cords. Many studies have revealed the therapeutic potential of NSCs. In this review, we discuss from an integrated view how NSCs can help SCI repair. We will discuss the sources and therapeutic potential of NSCs, as well as representative pre-clinical studies and clinical trials of NSC-based therapies for SCI repair.
Collapse
Affiliation(s)
- Wen Guo
- Department of Orthopaedic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Xindan Zhang
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, China
| | - Jiliang Zhai
- Department of Orthopaedic Surgery, Peking Union Medical College Hospital, Beijing, China
- *Correspondence: Jiliang Zhai, ; Jiajia Xue,
| | - Jiajia Xue
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, China
- *Correspondence: Jiliang Zhai, ; Jiajia Xue,
| |
Collapse
|
14
|
NPC transplantation rescues sci-driven cAMP/EPAC2 alterations, leading to neuroprotection and microglial modulation. Cell Mol Life Sci 2022; 79:455. [PMID: 35904607 PMCID: PMC9338125 DOI: 10.1007/s00018-022-04494-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 07/07/2022] [Accepted: 07/17/2022] [Indexed: 11/17/2022]
Abstract
Neural progenitor cell (NPC) transplantation represents a promising treatment strategy for spinal cord injury (SCI); however, the underlying therapeutic mechanisms remain incompletely understood. We demonstrate that severe spinal contusion in adult rats causes transcriptional dysregulation, which persists from early subacute to chronic stages of SCI and affects nearly 20,000 genes in total tissue extracts. Functional analysis of this dysregulated transcriptome reveals the significant downregulation of cAMP signalling components immediately after SCI, involving genes such as EPAC2 (exchange protein directly activated by cAMP), PKA, BDNF, and CAMKK2. The ectopic transplantation of spinal cord-derived NPCs at acute or subacute stages of SCI induces a significant transcriptional impact in spinal tissue, as evidenced by the normalized expression of a large proportion of SCI-affected genes. The transcriptional modulation pattern driven by NPC transplantation includes the rescued expression of cAMP signalling genes, including EPAC2. We also explore how the sustained in vivo inhibition of EPAC2 downstream signalling via the intrathecal administration of ESI-05 for 1 week impacts therapeutic mechanisms involved in the NPC-mediated treatment of SCI. NPC transplantation in SCI rats in the presence and absence of ESI-05 administration prompts increased rostral cAMP levels; however, NPC and ESI-05 treated animals exhibit a significant reduction in EPAC2 mRNA levels compared to animals receiving only NPCs treatment. Compared with transplanted animals, NPCs + ESI-05 treatment increases the scar area (as shown by GFAP staining), polarizes microglia into an inflammatory phenotype, and increases the magnitude of the gap between NeuN + cells across the lesion. Overall, our results indicate that the NPC-associated therapeutic mechanisms in the context of SCI involve the cAMP pathway, which reduces inflammation and provides a more neuropermissive environment through an EPAC2-dependent mechanism.
Collapse
|
15
|
Dermitzakis I, Manthou ME, Meditskou S, Miliaras D, Kesidou E, Boziki M, Petratos S, Grigoriadis N, Theotokis P. Developmental Cues and Molecular Drivers in Myelinogenesis: Revisiting Early Life to Re-Evaluate the Integrity of CNS Myelin. Curr Issues Mol Biol 2022; 44:3208-3237. [PMID: 35877446 PMCID: PMC9324160 DOI: 10.3390/cimb44070222] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/14/2022] [Accepted: 07/17/2022] [Indexed: 02/07/2023] Open
Abstract
The mammalian central nervous system (CNS) coordinates its communication through saltatory conduction, facilitated by myelin-forming oligodendrocytes (OLs). Despite the fact that neurogenesis from stem cell niches has caught the majority of attention in recent years, oligodendrogenesis and, more specifically, the molecular underpinnings behind OL-dependent myelinogenesis, remain largely unknown. In this comprehensive review, we determine the developmental cues and molecular drivers which regulate normal myelination both at the prenatal and postnatal periods. We have indexed the individual stages of myelinogenesis sequentially; from the initiation of oligodendrocyte precursor cells, including migration and proliferation, to first contact with the axon that enlists positive and negative regulators for myelination, until the ultimate maintenance of the axon ensheathment and myelin growth. Here, we highlight multiple developmental pathways that are key to successful myelin formation and define the molecular pathways that can potentially be targets for pharmacological interventions in a variety of neurological disorders that exhibit demyelination.
Collapse
Affiliation(s)
- Iasonas Dermitzakis
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.D.); (M.E.M.); (S.M.); (D.M.)
| | - Maria Eleni Manthou
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.D.); (M.E.M.); (S.M.); (D.M.)
| | - Soultana Meditskou
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.D.); (M.E.M.); (S.M.); (D.M.)
| | - Dimosthenis Miliaras
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.D.); (M.E.M.); (S.M.); (D.M.)
| | - Evangelia Kesidou
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, 54621 Thessaloniki, Greece; (E.K.); (M.B.); (N.G.)
| | - Marina Boziki
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, 54621 Thessaloniki, Greece; (E.K.); (M.B.); (N.G.)
| | - Steven Petratos
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC 3004, Australia;
| | - Nikolaos Grigoriadis
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, 54621 Thessaloniki, Greece; (E.K.); (M.B.); (N.G.)
| | - Paschalis Theotokis
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.D.); (M.E.M.); (S.M.); (D.M.)
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, 54621 Thessaloniki, Greece; (E.K.); (M.B.); (N.G.)
- Correspondence:
| |
Collapse
|
16
|
Bagheri Z, Shamsi F, Zeraatpisheh Z, Salmannejad M, Soltani A, Aligholi H. The Effect of Different Concentrations of Methylprednisolone on Survival, Proliferation, and Migration of Neural Stem/Progenitor Cells. Basic Clin Neurosci 2022; 13:207-213. [PMID: 36425947 PMCID: PMC9682315 DOI: 10.32598/bcn.2021.35.4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 04/13/2020] [Accepted: 07/26/2020] [Indexed: 08/25/2024] Open
Abstract
INTRODUCTION The present study addressed whether methylprednisolone (MP) as an anti-inflammatory drug used in neurodegenerative diseases and neural stem/progenitor cells (NS/PCs) is safe. METHODS First, embryonic rat NS/PCs were exposed to different concentrations of MP, and then we evaluated their survival by MTT assay, proliferation by analyzing the number and diameter of neurospheres, and the migration of the cells by neurosphere assay. RESULTS The viability of NS/PCs was reduced following exposure to 10, 15, and 20 μg/mL of MP. In addition, although the number of neurospheres did not change, exposure to different concentrations of MP resulted in the formation of smaller neurospheres. Despite these undesirable effects, the highest concentration of MP (20 μg/mL) increased the migration capacity of the NS/PCs. CONCLUSION The combination of MP and NS/PCs is not recommended due to the adverse effects of MP on the survival and proliferation of NS/PCs. HIGHLIGHTS Methylprednisolone reduced survival of neural stem/progenitor cells.Methylprednisolone decreased proliferation of neural stem/progenitor cells.The highest concentration of MP (20 μg/mL) increased the migration capacity of the neural stem/progenitor cells. PLAIN LANGUAGE SUMMARY In this study, we evaluate the effect of the exposure of neural stem/progenitor cells to methylprednisolone. Based on the results, combination of neural stem/progenitor cells and methylprednisolone not recommended due to reduction of survival and proliferation of the cells.
Collapse
Affiliation(s)
- Zohreh Bagheri
- Department of Physiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Shamsi
- Department of Neurosciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Neuroscience Laboratory (Brain, Cognition and Behavior), Department of Neurosciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Zeraatpisheh
- Department of Neurosciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Neuroscience Laboratory (Brain, Cognition and Behavior), Department of Neurosciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahin Salmannejad
- Department of Anatomy, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ahmad Soltani
- Department of Neurosurgery, Namazi Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hadi Aligholi
- Department of Neurosciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Neuroscience Laboratory (Brain, Cognition and Behavior), Department of Neurosciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Epilepsy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
17
|
Long-Term Effects of Neural Precursor Cell Transplantation on Secondary Injury Processes and Functional Recovery after Severe Cervical Contusion-Compression Spinal Cord Injury. Int J Mol Sci 2021; 22:ijms222313106. [PMID: 34884911 PMCID: PMC8658203 DOI: 10.3390/ijms222313106] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 11/29/2021] [Accepted: 12/02/2021] [Indexed: 01/21/2023] Open
Abstract
Cervical spinal cord injury (SCI) remains a devastating event without adequate treatment options despite decades of research. In this context, the usefulness of common preclinical SCI models has been criticized. We, therefore, aimed to use a clinically relevant animal model of severe cervical SCI to assess the long-term effects of neural precursor cell (NPC) transplantation on secondary injury processes and functional recovery. To this end, we performed a clip contusion-compression injury at the C6 level in 40 female Wistar rats and a sham surgery in 10 female Wistar rats. NPCs, isolated from the subventricular zone of green fluorescent protein (GFP) expressing transgenic rat embryos, were transplanted ten days after the injury. Functional recovery was assessed weekly, and FluoroGold (FG) retrograde fiber-labeling, as well as manganese-enhanced magnetic resonance imaging (MEMRI), were performed prior to the sacrifice of the animals eight weeks after SCI. After cryosectioning of the spinal cords, immunofluorescence staining was conducted. Results were compared between the treatment groups (NPC, Vehicle, Sham) and statistically analyzed (p < 0.05 was considered significant). Despite the severity of the injury, leading to substantial morbidity and mortality during the experiment, long-term survival of the engrafted NPCs with a predominant differentiation into oligodendrocytes could be observed after eight weeks. While myelination of the injured spinal cord was not significantly improved, NPC treated animals showed a significant increase of intact perilesional motor neurons and preserved spinal tracts compared to untreated Vehicle animals. These findings were associated with enhanced preservation of intact spinal cord tissue. However, reactive astrogliosis and inflammation where not significantly reduced by the NPC-treatment. While differences in the Basso–Beattie–Bresnahan (BBB) score and the Gridwalk test remained insignificant, animals in the NPC group performed significantly better in the more objective CatWalk XT gait analysis, suggesting some beneficial effects of the engrafted NPCs on the functional recovery after severe cervical SCI.
Collapse
|
18
|
Luo J, Shi X, Li L, Tan Z, Feng F, Li J, Pang M, Wang X, He L. An injectable and self-healing hydrogel with controlled release of curcumin to repair spinal cord injury. Bioact Mater 2021; 6:4816-4829. [PMID: 34136725 PMCID: PMC8175285 DOI: 10.1016/j.bioactmat.2021.05.022] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 05/08/2021] [Accepted: 05/08/2021] [Indexed: 12/16/2022] Open
Abstract
The harsh local micro-environment following spinal cord injury (SCI) remains a great challenge for neural regeneration. Local reconstitution of a favorable micro-environment by biocompatible scaffolds with desirable functions has thus been an area of concern. Herein, a hybrid hydrogel was developed using Fmoc-grafted chitosan (FC) and Fmoc peptide (FI). Dynamic reversible π-π stacking interactions of the fluorenyl rings enabled the FC/FI hybrid hydrogel to exhibit excellent injectable and self-healing properties, as characterized by visual appearances and rheological tests. Furthermore, the FC/FI hybrid hydrogel showed a slow and persistent release of curcumin (Cur), which was named as FC/FI-Cur hydrogel. In vitro studies confirmed that with the support of FC/FI-Cur hydrogel, neurite outgrowth was promoted, and Schwann cell (SC) migration away from dorsal root ganglia (DRG) spheres with enhanced myelination was substantiated. The FC/FI-Cur hydrogel well reassembled extracellular matrix at the lesion site of rat spinal cord and exerted outstanding effects in modulating local inflammatory reaction by regulating the phenotypes of infiltrated inflammatory cells. In addition, endogenous SCs were recruited in the FC/FI-Cur graft and participated in the remyelination process of the regenerated nerves. These outcomes favored functional recovery, as evidenced by improved hind limbs movement and enhanced electrophysiological properties. Thus, our study not only advanced the development of multifunctional hydrogels but also provided insights into comprehensive approaches for SCI repair.
Collapse
Affiliation(s)
- Jinghua Luo
- MOE Joint International Research Laboratory of CNS Regeneration, Jinan University, Guangzhou, 510632, China
- College of Life Science and Technology, Jinan University, Guangzhou, 510630, China
| | - Xueshuang Shi
- MOE Joint International Research Laboratory of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Liming Li
- MOE Joint International Research Laboratory of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Zan Tan
- Department of Spine Surgery, The 3rd Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Feng Feng
- Department of Spine Surgery, The 3rd Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Jun Li
- MOE Joint International Research Laboratory of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Mao Pang
- Department of Spine Surgery, The 3rd Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
| | - Xiaoying Wang
- College of Life Science and Technology, Jinan University, Guangzhou, 510630, China
| | - Liumin He
- MOE Joint International Research Laboratory of CNS Regeneration, Jinan University, Guangzhou, 510632, China
- Department of Spine Surgery, The 3rd Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China
- College of Life Science and Technology, Jinan University, Guangzhou, 510630, China
| |
Collapse
|
19
|
Zawadzka M, Kwaśniewska A, Miazga K, Sławińska U. Perspectives in the Cell-Based Therapies of Various Aspects of the Spinal Cord Injury-Associated Pathologies: Lessons from the Animal Models. Cells 2021; 10:cells10112995. [PMID: 34831217 PMCID: PMC8616284 DOI: 10.3390/cells10112995] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/25/2021] [Accepted: 10/31/2021] [Indexed: 02/07/2023] Open
Abstract
Traumatic injury of the spinal cord (SCI) is a devastating neurological condition often leading to severe dysfunctions, therefore an improvement in clinical treatment for SCI patients is urgently needed. The potential benefits of transplantation of various cell types into the injured spinal cord have been intensively investigated in preclinical SCI models and clinical trials. Despite the many challenges that are still ahead, cell transplantation alone or in combination with other factors, such as artificial matrices, seems to be the most promising perspective. Here, we reviewed recent advances in cell-based experimental strategies supporting or restoring the function of the injured spinal cord with a particular focus on the regenerative mechanisms that could define their clinical translation.
Collapse
|
20
|
Shao R, Zhang L, Yang H, Wang Y, Zhang Z, Yue J, Chen Y, Pan H, Zhou H, Quan R. Autophagy activation promotes the effect of iPSCs-derived NSCs on bladder function restoration after spinal cord injury. Tissue Cell 2021; 72:101596. [PMID: 34333229 DOI: 10.1016/j.tice.2021.101596] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 07/09/2021] [Accepted: 07/17/2021] [Indexed: 02/01/2023]
Abstract
The role of autophagy in the transplantation of induced pluripotent stem cells (iPSCs)-derived neural stem cells (NSCs) to treat spinal cord injury (SCI) and neurogenic bladder was investigated in this study. NSCs derived from human iPSCs were identified by and immunofluorescence assay. To clarify the role of autophagy, iPSCs were treated with either an autophagy inducer (rapamycin), or an autophagy inhibitor (chloroquine). Cell Counting kit-8 (CCK-8), western blot and flow cytometry were used to detect the effect of autophagy on the viability and differentiation of iPSCs. Sixty Wistar rats were selected to establish the SCI model and treated with iPSCs-derived NSCs transplantation. The effect of autophagy on the bladder function of rats with different treatments was evaluated by Basso, Beattie, and Bresnahan (BBB) score, bladder function score, bladder weight measurement, Hematoxylin & Eosin (H&E) staining, and Masson staining. The results of in vitro experiment showed that rapamycin enhanced the cell activity of iPSCs, increased the number of nestin positive cells, up-regulated Beclin-1 and LC3BI/II expressions, and down-regulated p62 expression. And the results of in vivo experiment showed that rapamycin improved exercise ability and bladder function, partially restored bladder weight, and significantly reduced bladder tissue damage in SCI rats. However, chloroquine showed the opposite results. The differentiation of iPSCs into NSCs could be promoted by induced autophagy, while neurogenic bladder of SCI was restored by autophagy activation.
Collapse
Affiliation(s)
- Rongxue Shao
- Department of Orthopedics, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, China.
| | - Liang Zhang
- Department of Orthopedics, Hangzhou Hospital of Traditional Chinese Medicine, China
| | - Hejie Yang
- Department of Orthopedics, Hangzhou Hospital of Traditional Chinese Medicine, China
| | - Yanbin Wang
- Department of Urology, The Third People's Hospital of Hangzhou, China
| | - Zhijing Zhang
- Department of Orthopedics, Hangzhou Hospital of Traditional Chinese Medicine, China
| | - Jun Yue
- Department of Orthopedics, Hangzhou Hospital of Traditional Chinese Medicine, China
| | - Yipeng Chen
- Research Institute of Orthopedics, Zhejiang Chinese Medical University, China
| | - Hao Pan
- Department of Orthopedics, Hangzhou Hospital of Traditional Chinese Medicine, China
| | - Hui Zhou
- Department of Orthopedics, Hangzhou Hospital of Traditional Chinese Medicine, China
| | - Renfu Quan
- Department of Orthopedics, Jiangnan Hospital Affiliated to Zhejiang Chinese Medical University, China
| |
Collapse
|
21
|
Feng J, Zhang Y, Zhu Z, Gu C, Waqas A, Chen L. Emerging Exosomes and Exosomal MiRNAs in Spinal Cord Injury. Front Cell Dev Biol 2021; 9:703989. [PMID: 34307384 PMCID: PMC8299525 DOI: 10.3389/fcell.2021.703989] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 06/18/2021] [Indexed: 12/16/2022] Open
Abstract
Acute spinal cord injury (SCI) is a serious traumatic event to the spinal cord with considerable morbidity and mortality. This injury leads to short- and long-term variations in the spinal cord, and can have a serious effect on the patient's sensory, motor, or autonomic functions. Due to the complicated pathological process of SCI, there is currently no successful clinical treatment strategy. Exosomes, extracellular vesicles (EVs) with a double-layer membrane structure of 30-150 nm diameter, have recently been considered as critical mediators for communication between cells and tissues by transferring proteins, lipids, and nucleic acids. Further studies verified that exosomes participate in the pathophysiological process of several diseases, including cancer, neurodegenerative diseases, and cardiovascular diseases, and could have a significant impact in their treatment. As natural carriers of biologically active cargos, exosomes have emerged as pathological mediators of SCI. In this review article, we critically discuss the functions of exosomes as intracellular mediators and potential treatments in SCI and provide an outlook on future research.
Collapse
Affiliation(s)
- Jia Feng
- Department of Neurosurgery, Neuroscience Center, Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yifan Zhang
- Department of Neurosurgery, Neuroscience Center, Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Zhihan Zhu
- School of Medicine, Southeast University, Nanjing, China
| | - Chenyang Gu
- Department of Neurosurgery, Neuroscience Center, Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Ahmed Waqas
- School of Medicine, Southeast University, Nanjing, China
| | - Lukui Chen
- Department of Neurosurgery, Neuroscience Center, Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
22
|
McIntyre WB, Pieczonka K, Khazaei M, Fehlings MG. Regenerative replacement of neural cells for treatment of spinal cord injury. Expert Opin Biol Ther 2021; 21:1411-1427. [PMID: 33830863 DOI: 10.1080/14712598.2021.1914582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Introduction: Traumatic Spinal Cord Injury (SCI) results from primary physical injury to the spinal cord, which initiates a secondary cascade of neural cell death. Current therapeutic approaches can attenuate the consequences of the primary and secondary events, but do not address the degenerative aspects of SCI. Transplantation of neural stem/progenitor cells (NPCs) for the replacement of the lost/damaged neural cells is suggested here as a regenerative approach that is complementary to current therapeutics.Areas Covered: This review addresses how neurons, oligodendrocytes, and astrocytes are impacted by traumatic SCI, and how current research in regenerative-NPC therapeutics aims to restore their functionality. Methods used to enhance graft survival, as well as bias progenitor cells towards neuronal, oligodendrogenic, and astroglia lineages are discussed.Expert Opinion: Despite an NPC's ability to differentiate into neurons, oligodendrocytes, and astrocytes in the transplant environment, their potential therapeutic efficacy requires further optimization prior to translation into the clinic. Considering the temporospatial identity of NPCs could promote neural repair in region specific injuries throughout the spinal cord. Moreover, understanding which cells are targeted by NPC-derived myelinating cells can help restore physiologically-relevant myelin patterns. Finally, the duality of astrocytes is discussed, outlining their context-dependent importance in the treatment of SCI.
Collapse
Affiliation(s)
- William Brett McIntyre
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada.,Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - Katarzyna Pieczonka
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada.,Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - Mohamad Khazaei
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Michael G Fehlings
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada.,Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada.,Department of Surgery, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
23
|
Fiani B, Jarrah R, Cathel A, Sarhadi K, Covarrubias C, Soula M. The role of gene therapy as a valuable treatment modality for multiple spinal pathologies. Regen Med 2021; 16:175-188. [PMID: 33709797 DOI: 10.2217/rme-2020-0147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The world of biomedical research has led to several breakthroughs in the treatment of various spinal pathologies. As we investigate chronic pathologies of the spine, we start to unravel the underlying molecular mechanisms through a careful analysis of mutated genetic sequences. Investigations have led to gene therapy being explored for its potential as a treatment modality. Despite only about 2% of current gene therapy trials being centered for spinal pathologies, spinal diseases are valuable targets in gene therapy administration. Through a comprehensive literature review, our objective is to discuss the molecular mechanisms behind gene therapy for spinal pathologies, the genetic targets, along with the outcomes, success, and possible pitfalls in gene therapy research and administration. The emerging development of robotic technologies and intelligent carriers are recognized as a promising innovative technique for increasing the efficiency of gene therapy and potentially resolving spinal pathologies.
Collapse
Affiliation(s)
- Brian Fiani
- Department of Neurosurgery, Desert Regional Medical Center, Palm Springs, CA 92262, USA
| | - Ryan Jarrah
- College of Arts & Science, University of Michigan-Flint, Flint, MI 48502, USA
| | - Alessandra Cathel
- Department of Neurosurgery, Desert Regional Medical Center, Palm Springs, CA 92262, USA
| | - Kasra Sarhadi
- Department of Neurology, University of Washington, Seattle, WA 98195, USA
| | - Claudia Covarrubias
- School of Medicine, Universidad Anáhuac Querétaro, Santiago de Querétaro 76246, México
| | - Marisol Soula
- School of Medicine, New York University, NY 10016, USA
| |
Collapse
|
24
|
Abstract
Traumatic spinal cord injury (SCI) results in direct and indirect damage to neural tissues, which results in motor and sensory dysfunction, dystonia, and pathological reflex that ultimately lead to paraplegia or tetraplegia. A loss of cells, axon regeneration failure, and time-sensitive pathophysiology make tissue repair difficult. Despite various medical developments, there are currently no effective regenerative treatments. Stem cell therapy is a promising treatment for SCI due to its multiple targets and reactivity benefits. The present review focuses on SCI stem cell therapy, including bone marrow mesenchymal stem cells, umbilical mesenchymal stem cells, adipose-derived mesenchymal stem cells, neural stem cells, neural progenitor cells, embryonic stem cells, induced pluripotent stem cells, and extracellular vesicles. Each cell type targets certain features of SCI pathology and shows therapeutic effects via cell replacement, nutritional support, scaffolds, and immunomodulation mechanisms. However, many preclinical studies and a growing number of clinical trials found that single-cell treatments had only limited benefits for SCI. SCI damage is multifaceted, and there is a growing consensus that a combined treatment is needed.
Collapse
Affiliation(s)
- Liyi Huang
- Department of Rehabilitation Medicine Center, 34753West China Hospital/West China School of Medicine, Sichuan University, Chengdu, Sichuan, PR China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Sichuan University, Chengdu, Sichuan Province, PR China
| | - Chenying Fu
- State Key Laboratory of Biotherapy, 34753West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Feng Xiong
- Department of Rehabilitation Medicine Center, 34753West China Hospital/West China School of Medicine, Sichuan University, Chengdu, Sichuan, PR China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Sichuan University, Chengdu, Sichuan Province, PR China
| | - Chengqi He
- Department of Rehabilitation Medicine Center, 34753West China Hospital/West China School of Medicine, Sichuan University, Chengdu, Sichuan, PR China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Sichuan University, Chengdu, Sichuan Province, PR China
| | - Quan Wei
- Department of Rehabilitation Medicine Center, 34753West China Hospital/West China School of Medicine, Sichuan University, Chengdu, Sichuan, PR China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Sichuan University, Chengdu, Sichuan Province, PR China
| |
Collapse
|
25
|
Jones I, Novikova LN, Wiberg M, Carlsson L, Novikov LN. Human Embryonic Stem Cell-derived Neural Crest Cells Promote Sprouting and Motor Recovery Following Spinal Cord Injury in Adult Rats. Cell Transplant 2021; 30:963689720988245. [PMID: 33522309 PMCID: PMC7863557 DOI: 10.1177/0963689720988245] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Spinal cord injury results in irreversible tissue damage and permanent sensorimotor impairment. The development of novel therapeutic strategies that improve the life quality of affected individuals is therefore of paramount importance. Cell transplantation is a promising approach for spinal cord injury treatment and the present study assesses the efficacy of human embryonic stem cell–derived neural crest cells as preclinical cell-based therapy candidates. The differentiated neural crest cells exhibited characteristic molecular signatures and produced a range of biologically active trophic factors that stimulated in vitro neurite outgrowth of rat primary dorsal root ganglia neurons. Transplantation of the neural crest cells into both acute and chronic rat cervical spinal cord injury models promoted remodeling of descending raphespinal projections and contributed to the partial recovery of forelimb motor function. The results achieved in this proof-of-concept study demonstrates that human embryonic stem cell–derived neural crest cells warrant further investigation as cell-based therapy candidates for the treatment of spinal cord injury.
Collapse
Affiliation(s)
- Iwan Jones
- 59588Umeå Center for Molecular Medicine, Umeå University, Umeå, Sweden.,Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | | | - Mikael Wiberg
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden.,Department of Surgical and Perioperative Science, Section of Hand and Plastic Surgery, Umeå University, Umeå, Sweden
| | - Leif Carlsson
- 59588Umeå Center for Molecular Medicine, Umeå University, Umeå, Sweden
| | - Lev N Novikov
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| |
Collapse
|
26
|
Li Y, Shen PP, Wang B. Induced pluripotent stem cell technology for spinal cord injury: a promising alternative therapy. Neural Regen Res 2021; 16:1500-1509. [PMID: 33433463 PMCID: PMC8323703 DOI: 10.4103/1673-5374.303013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Spinal cord injury has long been a prominent challenge in the trauma repair process. Spinal cord injury is a research hotspot by virtue of its difficulty to treat and its escalating morbidity. Furthermore, spinal cord injury has a long period of disease progression and leads to complications that exert a lot of mental and economic pressure on patients. There are currently a large number of therapeutic strategies for treating spinal cord injury, which range from pharmacological and surgical methods to cell therapy and rehabilitation training. All of these strategies have positive effects in the course of spinal cord injury treatment. This review mainly discusses the problems regarding stem cell therapy for spinal cord injury, including the characteristics and action modes of all relevant cell types. Induced pluripotent stem cells, which represent a special kind of stem cell population, have gained impetus in cell therapy development because of a range of advantages. Induced pluripotent stem cells can be developed into the precursor cells of each neural cell type at the site of spinal cord injury, and have great potential for application in spinal cord injury therapy.
Collapse
Affiliation(s)
- Yu Li
- Clinical Stem Cell Center, the Affiliated Drum Tower Hospital of Nanjing University Medical School, School of Life Science, Nanjing University, Nanjing, Jiangsu Province, China
| | - Ping-Ping Shen
- State Key Laboratory of Pharmaceutical Biotechnology and The Comprehensive Cancer Center, the Affiliated Drum Tower Hospital of Nanjing University Medical School, School of Life Science, Nanjing University, Nanjing, Jiangsu Province, China
| | - Bin Wang
- Clinical Stem Cell Center, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China
| |
Collapse
|
27
|
Vancamp P, Butruille L, Demeneix BA, Remaud S. Thyroid Hormone and Neural Stem Cells: Repair Potential Following Brain and Spinal Cord Injury. Front Neurosci 2020; 14:875. [PMID: 32982671 PMCID: PMC7479247 DOI: 10.3389/fnins.2020.00875] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 07/28/2020] [Indexed: 12/22/2022] Open
Abstract
Neurodegenerative diseases are characterized by chronic neuronal and/or glial cell loss, while traumatic injury is often accompanied by the acute loss of both. Multipotent neural stem cells (NSCs) in the adult mammalian brain spontaneously proliferate, forming neuronal and glial progenitors that migrate toward lesion sites upon injury. However, they fail to replace neurons and glial cells due to molecular inhibition and the lack of pro-regenerative cues. A major challenge in regenerative biology therefore is to unveil signaling pathways that could override molecular brakes and boost endogenous repair. In physiological conditions, thyroid hormone (TH) acts on NSC commitment in the subventricular zone, and the subgranular zone, the two largest NSC niches in mammals, including humans. Here, we discuss whether TH could have beneficial actions in various pathological contexts too, by evaluating recent data obtained in mammalian models of multiple sclerosis (MS; loss of oligodendroglial cells), Alzheimer’s disease (loss of neuronal cells), stroke and spinal cord injury (neuroglial cell loss). So far, TH has shown promising effects as a stimulator of remyelination in MS models, while its role in NSC-mediated repair in other diseases remains elusive. Disentangling the spatiotemporal aspects of the injury-driven repair response as well as the molecular and cellular mechanisms by which TH acts, could unveil new ways to further exploit its pro-regenerative potential, while TH (ant)agonists with cell type-specific action could provide safer and more target-directed approaches that translate easier to clinical settings.
Collapse
Affiliation(s)
- Pieter Vancamp
- Laboratory Molecular Physiology and Adaptation, CNRS UMR 7221, Muséum National d'Histoire Naturelle, Department Adaptations of Life, Paris, France
| | - Lucile Butruille
- Laboratory Molecular Physiology and Adaptation, CNRS UMR 7221, Muséum National d'Histoire Naturelle, Department Adaptations of Life, Paris, France
| | - Barbara A Demeneix
- Laboratory Molecular Physiology and Adaptation, CNRS UMR 7221, Muséum National d'Histoire Naturelle, Department Adaptations of Life, Paris, France
| | - Sylvie Remaud
- Laboratory Molecular Physiology and Adaptation, CNRS UMR 7221, Muséum National d'Histoire Naturelle, Department Adaptations of Life, Paris, France
| |
Collapse
|
28
|
Ma Y, Deng M, Zhao XQ, Liu M. Alternatively Polarized Macrophages Regulate the Growth and Differentiation of Ependymal Stem Cells through the SIRT2 Pathway. Exp Neurobiol 2020; 29:150-163. [PMID: 32408405 PMCID: PMC7237271 DOI: 10.5607/en19078] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 04/15/2020] [Accepted: 04/16/2020] [Indexed: 02/06/2023] Open
Abstract
Ependymal stem cells (EpSCs) are dormant stem cells in the adult spinal cord that proliferate rapidly and migrate to the site of injury after spinal cord injury (SCI). Although they can differentiate into neurons under appropriate conditions in vitro, EpSCs mainly differentiate into astrocytes in vivo. Our previous study confirmed that alternatively polarized macrophages (M2) facilitate the differentiation of EpSCs towards neurons, but the detailed mechanism remains elusive. In the present study, we found that M2 conditioned medium could upregulate the expression of Sirtuin 2 (SIRT2) in EpSCs in vitro through the BDNF/TrkB-MEK/ERK signaling pathway. As an important deacetylase, SIRT2 deacetylated stable Ac-α-tubulin (Acetyl alpha Tubulin) in microtubules and thus promoted EpSC differentiation into neurons. The present study provides a theoretical basis and a new way to improve neural recovery, such as regulating the growth and differentiation of EpSCs by increasing the proportion of M2 cells in the local microenvironment or upregulating the expression of SIRT2 in EpSCs.
Collapse
Affiliation(s)
- Yonggang Ma
- Department of Orthopaedics, Renmin Hospital, Wuhan University, Wuhan 430060, China
| | - Ming Deng
- Department of Orthopaedics, Renmin Hospital, Wuhan University, Wuhan 430060, China
| | - Xiao-Qi Zhao
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Min Liu
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| |
Collapse
|
29
|
Ottoboni L, von Wunster B, Martino G. Therapeutic Plasticity of Neural Stem Cells. Front Neurol 2020; 11:148. [PMID: 32265815 PMCID: PMC7100551 DOI: 10.3389/fneur.2020.00148] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 02/14/2020] [Indexed: 12/21/2022] Open
Abstract
Neural stem cells (NSCs) have garnered significant scientific and commercial interest in the last 15 years. Given their plasticity, defined as the ability to develop into different phenotypes inside and outside of the nervous system, with a capacity of almost unlimited self-renewal, of releasing trophic and immunomodulatory factors, and of exploiting temporal and spatial dynamics, NSCs have been proposed for (i) neurotoxicity testing; (ii) cellular therapies to treat CNS diseases; (iii) neural tissue engineering and repair; (iv) drug target validation and testing; (v) personalized medicine. Moreover, given the growing interest in developing cell-based therapies to target neurodegenerative diseases, recent progress in developing NSCs from human-induced pluripotent stem cells has produced an analog of endogenous NSCs. Herein, we will review the current understanding on emerging conceptual and technological topics in the neural stem cell field, such as deep characterization of the human compartment, single-cell spatial-temporal dynamics, reprogramming from somatic cells, and NSC manipulation and monitoring. Together, these aspects contribute to further disentangling NSC plasticity to better exploit the potential of those cells, which, in the future, might offer new strategies for brain therapies.
Collapse
Affiliation(s)
- Linda Ottoboni
- Neurology and Neuroimmunology Unit, Institute of Experimental Neurology, San Raffaele Scientific Institute, Milan, Italy
| | | | - Gianvito Martino
- Neurology and Neuroimmunology Unit, Institute of Experimental Neurology, San Raffaele Scientific Institute, Milan, Italy.,Università Vita-Salute San Raffaele, School of Medicine, Milan, Italy
| |
Collapse
|
30
|
Modulating Pro-inflammatory Cytokines, Tissue Damage Magnitude, and Motor Deficit in Spinal Cord Injury with Subventricular Zone-Derived Extracellular Vesicles. J Mol Neurosci 2019; 70:458-466. [DOI: 10.1007/s12031-019-01437-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 11/04/2019] [Indexed: 02/06/2023]
|