1
|
Wang NB, Adewumi HO, Lende-Dorn BA, Beitz AM, O'Shea TM, Galloway KE. Compact transcription factor cassettes generate functional, engraftable motor neurons by direct conversion. Cell Syst 2025; 16:101206. [PMID: 40086435 DOI: 10.1016/j.cels.2025.101206] [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: 12/28/2023] [Revised: 11/07/2024] [Accepted: 02/11/2025] [Indexed: 03/16/2025]
Abstract
Direct conversion generates patient-specific, disease-relevant cell types, such as neurons, that are rare, limited, or difficult to isolate from common and easily accessible cells, such as skin cells. However, low rates of direct conversion and complex protocols limit scalability and, thus, the potential of cell-fate conversion for biomedical applications. Here, we optimize the conversion protocol by examining process parameters, including transcript design; delivery via adeno-associated virus (AAV), retrovirus, and lentivirus; cell seeding density; and the impact of media conditions. Thus, we report a compact, portable conversion process that boosts proliferation and increases direct conversion of mouse fibroblasts to induced motor neurons (iMNs) to achieve high conversion rates of above 1,000%, corresponding to more than ten motor neurons yielded per cell seeded, which we achieve through expansion. Our optimized, direct conversion process generates functional motor neurons at scales relevant for cell therapies (>107 cells) that graft with the mouse central nervous system. High-efficiency, compact, direct conversion systems will support scaling to patient-specific, neural cell therapies.
Collapse
Affiliation(s)
- Nathan B Wang
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Honour O Adewumi
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Brittany A Lende-Dorn
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Adam M Beitz
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Timothy M O'Shea
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Kate E Galloway
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
2
|
Heller RA, Maares M, Chillon TS, Witte H, Al-Halabi OT, Heene S, Younsi A, Haubruck P, Schomburg L, Moghaddam A, Biglari B, Haase H. Total and Free Zinc Dynamics as Biomarkers for Neurological Impairment in Traumatic Spinal Cord Injury. Nutrients 2025; 17:496. [PMID: 39940353 PMCID: PMC11820840 DOI: 10.3390/nu17030496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/15/2025] [Accepted: 01/24/2025] [Indexed: 02/16/2025] Open
Abstract
Background/Objectives: Traumatic spinal cord injury (TSCI) profoundly impacts patients by precipitating a loss of motor and sensory capabilities, largely due to oxidative stress and inflammation during the secondary injury phase. Methods: This investigation explores the diagnostic potential of zinc (Zn) and free zinc (fZn) as biomarkers by analyzing their serum concentration dynamics in 48 TSCI individuals with TSCI, with the aim of correlating these levels with neurological impairment. Serum samples collected at admission, 4 h, 9 h, 12 h, 24 h, and 3 days post-injury were analyzed for total serum Zn and fZn concentrations. The patients were compared to a control group comprised of individuals with vertebral fractures but no neurological deficits. Results: The study revealed injury-specific fluctuations in Zn and fZn levels following TSCI, with significantly lower Zn levels observed post-TSCI compared to controls (p = 0.016). The American Spinal Injury Association (ASIA) Impairment scale (AIS) assessments at admission and three months post-injury showed Zn level differences are linked to neurological recovery (AIS+:1 > AIS+:2, 0 h: p = 0.008; AIS+:0 < AIS+:1, 4 h: p = 0.016), highlighting the critical role of Zn and trace elements in the early remission process after TSCI. Notably, significant differences in fZn levels were detected between the control and TSCI groups (TSCI < Control; 12 h: p = 0.045; 24 h: p = 0.001; 3 d: p = 0.016), with the peak diagnostic performance of fZn at 24 h post-injury, as indicated by an Area Under the ROC Curve (AUC) of 83.84% (CI: 0.698-0.978). Conclusions: These findings underscore the potential of fZn as a biomarker to guide early diagnostic and therapeutic interventions aimed at mitigating secondary injury and enhancing recovery outcomes. This study contributes insights into the dynamics of serum Zn and its importance, holding specific diagnostic properties that could be critically relevant in the early phase of biomarker signature development for TSCI diagnostics and prognosis.
Collapse
Affiliation(s)
- Raban Arved Heller
- Institute for Experimental Endocrinology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie, Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, 13353 Berlin, Germany; (R.A.H.); (T.S.C.); (L.S.)
- Department of Food Chemistry and Toxicology, Technische Universität Berlin, Straße des 17. Juni 135, 10623 Berlin, Germany;
- Department of Traumatology and Orthopaedics, Septic and Reconstructive Surgery, Bundeswehr Hospital Berlin, 10115 Berlin, Germany
| | - Maria Maares
- Department of Food Chemistry and Toxicology, Technische Universität Berlin, Straße des 17. Juni 135, 10623 Berlin, Germany;
| | - Thilo Samson Chillon
- Institute for Experimental Endocrinology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie, Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, 13353 Berlin, Germany; (R.A.H.); (T.S.C.); (L.S.)
| | - Hanno Witte
- Department of Hematology and Oncology, Bundeswehr Hospital Ulm, 89081 Ulm, Germany;
| | - Obada T. Al-Halabi
- Department of Neurosurgery, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany; (O.T.A.-H.); (A.Y.)
- Medical Faculty, Heidelberg University, Grabengasse 1, 69117 Heidelberg, Germany
| | - Stefan Heene
- Department of Neurosurgery, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany; (O.T.A.-H.); (A.Y.)
- Medical Faculty, Heidelberg University, Grabengasse 1, 69117 Heidelberg, Germany
| | - Alexander Younsi
- Department of Neurosurgery, Heidelberg University Hospital, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany; (O.T.A.-H.); (A.Y.)
- Medical Faculty, Heidelberg University, Grabengasse 1, 69117 Heidelberg, Germany
| | - Patrick Haubruck
- Raymond-Purves Bone and Joint Research Laboratories, Kolling Institute of Medical Research, Institute of Bone and Joint Research, University of Sydney, St Leonards, NSW 2065, Australia;
| | - Lutz Schomburg
- Institute for Experimental Endocrinology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie, Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, 13353 Berlin, Germany; (R.A.H.); (T.S.C.); (L.S.)
| | - Arash Moghaddam
- Orthopedic and Trauma Surgery, Frohsinnstraße 12, 63739 Aschaffenburg, Germany
| | - Bahram Biglari
- Department of Paraplegiology, BG Trauma Centre Ludwigshafen, 67071 Ludwigshafen, Germany;
| | - Hajo Haase
- Department of Food Chemistry and Toxicology, Technische Universität Berlin, Straße des 17. Juni 135, 10623 Berlin, Germany;
| |
Collapse
|
3
|
Ma Z, Liu T, Liu L, Pei Y, Wang T, Wang Z, Guan Y, Zhang X, Zhang Y, Chen X. Epidermal Neural Crest Stem Cell Conditioned Medium Enhances Spinal Cord Injury Recovery via PI3K/AKT-Mediated Neuronal Apoptosis Suppression. Neurochem Res 2024; 49:2854-2870. [PMID: 39023805 PMCID: PMC11365850 DOI: 10.1007/s11064-024-04207-8] [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: 09/23/2023] [Revised: 04/19/2024] [Accepted: 07/05/2024] [Indexed: 07/20/2024]
Abstract
This study aimed to assess the impact of conditioned medium from epidermal neural crest stem cells (EPI-NCSCs-CM) on functional recovery following spinal cord injury (SCI), while also exploring the involvement of the PI3K-AKT signaling pathway in regulating neuronal apoptosis. EPI-NCSCs were isolated from 10-day-old Sprague-Dawley rats and cultured for 48 h to obtain EPI-NCSC-CM. SHSY-5Y cells were subjected with H2O2 treatment to induce apoptosis. Cell viability and survival rates were evaluated using the CCK-8 assay and calcein-AM/PI staining. SCI contusion model was established in adult Sprague-Dawley rats to assess functional recovery, utilizing the Basso, Beattie and Bresnahan (BBB) scoring system, inclined test, and footprint observation. Neurological restoration after SCI was analyzed through electrophysiological recordings. Histological analysis included hematoxylin and eosin (H&E) staining and Nissl staining to evaluate tissue organization. Apoptosis and oxidative stress levels were assessed using TUNEL staining and ROS detection methods. Additionally, western blotting was performed to examine the expression of apoptotic markers and proteins related to the PI3K/AKT signaling pathway. EPI-NCSC-CM significantly facilitated functional and histological recovery in SCI rats by inhibiting neuronal apoptosis through modulation of the PI3K/AKT pathway. Administration of EPI-NCSCs-CM alleviated H2O2-induced neurotoxicity in SHSY-5Y cells in vitro. The use of LY294002, a PI3K inhibitor, underscored the crucial role of the PI3K/AKT signaling pathway in regulating neuronal apoptosis. This study contributes to the ongoing exploration of molecular pathways involved in spinal cord injury (SCI) repair, focusing on the therapeutic potential of EPI-NCSC-CM. The research findings indicate that EPI-NCSC-CM exerts a neuroprotective effect by suppressing neuronal apoptosis through activation of the PI3K/AKT pathway in SCI rats. These results highlight the promising role of EPI-NCSC-CM as a potential treatment strategy for SCI, emphasizing the significance of the PI3K/AKT pathway in mediating its beneficial effects.
Collapse
Affiliation(s)
- Ziqian Ma
- Department of Orthopedics Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
- Department of Orthopedics, Beijing Chaoyang Hospital, Capital Medical University, 8 Workers Stadium South Road, Chaoyang District, Beijing, China
| | - Tao Liu
- Department of Orthopedics Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Liang Liu
- Department of Orthopedics Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Yilun Pei
- Department of Orthopedics Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Tianyi Wang
- Department of Orthopedics, 981st Hospital of the Chinese People's Liberation Army Joint Logistics Support Force, Chengde, 067000, Hebei Province, P.R. China
| | - Zhijie Wang
- Department of Pediatric Internal Medicine, Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Yun Guan
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
- Department of Neurological Surgery, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Xinwei Zhang
- Department of Orthopedics Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Yan Zhang
- Department of Orthopedics Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China.
| | - Xueming Chen
- Department of Orthopedics Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
4
|
Pu T, Wang J, Wei J, Zeng A, Zhang J, Chen J, Yin L, Li J, Lin TP, Melamed J, Corey E, Gao AC, Wu BJ. Stromal-derived MAOB promotes prostate cancer growth and progression. SCIENCE ADVANCES 2024; 10:eadi4935. [PMID: 38335292 PMCID: PMC10857382 DOI: 10.1126/sciadv.adi4935] [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: 04/28/2023] [Accepted: 01/09/2024] [Indexed: 02/12/2024]
Abstract
Prostate cancer (PC) develops in a microenvironment where the stromal cells modulate adjacent tumor growth and progression. Here, we demonstrated elevated levels of monoamine oxidase B (MAOB), a mitochondrial enzyme that degrades biogenic and dietary monoamines, in human PC stroma, which was associated with poor clinical outcomes of PC patients. Knockdown or overexpression of MAOB in human prostate stromal fibroblasts indicated that MAOB promotes cocultured PC cell proliferation, migration, and invasion and co-inoculated prostate tumor growth in mice. Mechanistically, MAOB induces a reactive stroma with activated marker expression, increased extracellular matrix remodeling, and acquisition of a protumorigenic phenotype through enhanced production of reactive oxygen species. Moreover, MAOB transcriptionally activates CXCL12 through Twist1 synergizing with TGFβ1-dependent Smads in prostate stroma, which stimulates tumor-expressed CXCR4-Src/JNK signaling in a paracrine manner. Pharmacological inhibition of stromal MAOB restricted PC xenograft growth in mice. Collectively, these findings characterize the contribution of MAOB to PC and suggest MAOB as a potential stroma-based therapeutic target.
Collapse
Affiliation(s)
- Tianjie Pu
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA
| | - Jing Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA
| | - Jing Wei
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA
| | - Alan Zeng
- Undergraduate Programs, University of Washington, Seattle, WA 98195, USA
| | - Jinglong Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA
| | - Jingrui Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA
| | - Lijuan Yin
- Uro-Oncology Research Program, Samuel Oschin Comprehensive Cancer Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jingjing Li
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA
| | - Tzu-Ping Lin
- Department of Urology, Taipei Veterans General Hospital, Taipei 11217, Taiwan, Republic of China
- Department of Urology, School of Medicine and Shu-Tien Urological Research Center, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan, Republic of China
| | - Jonathan Melamed
- Department of Pathology, Grossman School of Medicine, New York University, New York, NY 10016, USA
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, WA 98195, USA
| | - Allen C. Gao
- Department of Urologic Surgery, University of California, Davis, Sacramento, CA 95817, USA
| | - Boyang Jason Wu
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA
| |
Collapse
|
5
|
Nagaoki T, Kumagai G, Nitobe Y, Sasaki A, Fujita T, Fukutoku T, Saruta K, Tsukuda M, Asari T, Wada K, Dezawa M, Ishibashi Y. Comparison of the Anti-Inflammatory Effects of Mouse Adipose- and Bone-Marrow-Derived Multilineage-Differentiating Stress-Enduring Cells in Acute-Phase Spinal Cord Injury. J Neurotrauma 2023; 40:2596-2609. [PMID: 37051701 DOI: 10.1089/neu.2022.0470] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2023] Open
Abstract
Abstract Spinal cord injury (SCI) is a serious neurological disorder, with the consequent disabilities conferred by this disorder typically persisting for life. Multilineage-differentiating stress-enduring (Muse) cells are endogenous stem cells that can be collected from various tissues as well as from mesenchymal stem cells (MSCs); additionally, these Muse cells are currently being used in clinical trials. The anti-inflammatory effect of stem cell transplantation prevents secondary injuries of SCI; however, its effect on Muse cells remains unclear. In this study, we aimed to compare the anti-inflammatory effects of adipose (AD)- and bone marrow (BM)-Muse cells that were isolated from mice (6-week-old C57BL/6J) following intralesional administration during the acute phase of SCI. Flow cytometry was used to isolate Muse cells from AD and BM MSCs. The percentage of Muse cells was 3.9 and 2.7% for AD and BM MSCs, respectively. To examine cell viability, Muse cells were incubated under H2O2-induced oxidative stress conditions. Overall, AD-Muse cells exhibited higher viability than BM-Muse cells (p = 0.032). In enzyme-linked immunosorbent assay analysis, AD-Muse cells displayed greater secretion of brain-derived neurotrophic factor (BDNF; p = 0.008), vascular endothelial growth factor (p = 0.032), and hepatocyte growth factor (p = 0.016). DNA microarray analysis revealed higher expression of Bdnf, neurotrophin-3 (Ntf3), nerve growth factor (Ngf), pleiotrophin (Ptn), and midkine (Mdk) in AD-Muse cells than in BM-Muse cells. To assess their anti-inflammatory effects in vitro, Muse cells and macrophages were co-cultured, and the levels of cytokines (tumor necrosis factor [TNF] α and interleukin [IL] 10) were measured in the medium. Consequently, we found that TNFα levels were lower in AD-Muse cells than in BM-Muse cells (p = 0.009), and IL10 levels were higher in AD-Muse cells than in BM-Muse cells (p = 0.008). Further, we induced moderate injuries via contusion of the spinal cord at the T10 level; Muse cells were transplanted intralesionally 7 days post-SCI. The number of surviving cells, alongside the number of CD86+ (M1 inflammatory effect), and CD206+ (M2 anti-inflammatory effect) macrophages in the spinal cord were measured 7 days post-transplantation. The number of surviving AD-Muse cells was higher than the number of surviving BM-Muse cells (ratio of AD-Muse/BM-Muse = 2.5, p > 0.05). The M1/M2 ratio in the AD-Muse cell-group (0.37) was lower than that in the control (phosphate-buffered saline) group (3.60, p = 0.008). The lesion area in the AD-Muse cell group was smaller than that in the BM-non-Muse (p = 0.049) and control groups (p = 0.012). As AD-Muse cells conferred a higher cell survival and neurotrophic factor secretion ability in vitro, AD-Muse cells demonstrated reduced inflammation after SCI. Overall, intralesional AD-Muse cell therapy is a potential therapeutic candidate that is expected to exhibit anti-inflammatory effects following acute SCI.
Collapse
Affiliation(s)
- Toshihide Nagaoki
- Department of Orthopedic Surgery, Hirosaki University Graduate School of Medicine, Aomori, Japan
| | - Gentaro Kumagai
- Department of Orthopedic Surgery, Hirosaki University Graduate School of Medicine, Aomori, Japan
| | - Yohshiro Nitobe
- Department of Orthopedic Surgery, Hirosaki University Graduate School of Medicine, Aomori, Japan
| | - Ayako Sasaki
- Department of Orthopedic Surgery, Hirosaki University Graduate School of Medicine, Aomori, Japan
| | - Taku Fujita
- Department of Orthopedic Surgery, Hirosaki University Graduate School of Medicine, Aomori, Japan
| | - Tatsuhiro Fukutoku
- Department of Orthopedic Surgery, Hirosaki University Graduate School of Medicine, Aomori, Japan
| | - Kenya Saruta
- Department of Orthopedic Surgery, Hirosaki University Graduate School of Medicine, Aomori, Japan
| | - Manami Tsukuda
- Department of Orthopedic Surgery, Hirosaki University Graduate School of Medicine, Aomori, Japan
| | - Toru Asari
- Department of Orthopedic Surgery, Hirosaki University Graduate School of Medicine, Aomori, Japan
| | - Kanichiro Wada
- Department of Orthopedic Surgery, Hirosaki University Graduate School of Medicine, Aomori, Japan
| | - Mari Dezawa
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Yasuyuki Ishibashi
- Department of Orthopedic Surgery, Hirosaki University Graduate School of Medicine, Aomori, Japan
| |
Collapse
|
6
|
Huang L, Sun X, Wang L, Pei G, Wang Y, Zhang Q, Liang Z, Wang D, Fu C, He C, Wei Q. Enhanced effect of combining bone marrow mesenchymal stem cells (BMMSCs) and pulsed electromagnetic fields (PEMF) to promote recovery after spinal cord injury in mice. MedComm (Beijing) 2022; 3:e160. [PMID: 35949547 PMCID: PMC9350428 DOI: 10.1002/mco2.160] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 06/21/2022] [Accepted: 06/24/2022] [Indexed: 12/03/2022] Open
Abstract
Spinal cord injury (SCI) is a destructive traumatic disease of the central nervous system without satisfying therapy efficiency. Bone marrow mesenchymal stem cells (BMMSCs) therapy promotes the neurotrophic factors' secretion and axonal regeneration, thereby promoting recovery of SCI. Pulsed electromagnetic fields (PEMF) therapy has been proven to promote neural growth and regeneration. Both BMMSCs and PEMF have shown curative effects for SCI; PEMF can further promote stem cell differentiation. Thus, we explored the combined effects of BMMSCs and PEMF and the potential interaction between these two therapies in SCI. Compared with the SCI control, BMMSCs, and PEMF groups, the combinational therapy displayed the best therapeutic effect. Combinational therapy increased the expression levels of nutritional factors including brain-derived neurotrophic factor (BDNF), nerve growth factors (NGF) and vascular endothelial growth factor (VEGF), enhanced neuron preservation (NeuN and NF-200), and increased axonal growth (MBP and myelin sheath). Additionally, PEMF promoted the expression levels of BDNF and VEGF in BMMSCs via Wnt/β-catenin signaling pathway. In summary, the combined therapy of BMMSCs and PEMF displayed a more satisfactory effect than BMMSCs and PEMF therapy alone, indicating a promising application of combined therapy for the therapy of SCI.
Collapse
Affiliation(s)
- Liyi Huang
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China HospitalSichuan UniversityChengduPR China
- Key Laboratory of Rehabilitation Medicine in Sichuan ProvinceSichuan UniversityChengduPR China
| | - Xin Sun
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China HospitalSichuan UniversityChengduPR China
- Key Laboratory of Rehabilitation Medicine in Sichuan ProvinceSichuan UniversityChengduPR China
| | - Lu Wang
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China HospitalSichuan UniversityChengduPR China
- Key Laboratory of Rehabilitation Medicine in Sichuan ProvinceSichuan UniversityChengduPR China
| | - Gaiqing Pei
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China HospitalSichuan UniversityChengduPR China
- Key Laboratory of Rehabilitation Medicine in Sichuan ProvinceSichuan UniversityChengduPR China
| | - Yang Wang
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China HospitalSichuan UniversityChengduPR China
- Key Laboratory of Rehabilitation Medicine in Sichuan ProvinceSichuan UniversityChengduPR China
| | - Qing Zhang
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China HospitalSichuan UniversityChengduPR China
- Key Laboratory of Rehabilitation Medicine in Sichuan ProvinceSichuan UniversityChengduPR China
| | - Zejun Liang
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China HospitalSichuan UniversityChengduPR China
- Key Laboratory of Rehabilitation Medicine in Sichuan ProvinceSichuan UniversityChengduPR China
| | - Dong Wang
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China HospitalSichuan UniversityChengduPR China
- Key Laboratory of Rehabilitation Medicine in Sichuan ProvinceSichuan UniversityChengduPR China
| | - Chenying Fu
- National Clinical Research Center for Geriatrics, West China HospitalSichuan UniversityChengduSichuanPR China
- Aging and Geriatric Mechanism Laboratory, West China HospitalSichuan UniversityChengduSichuanPR China
| | - Chengqi He
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China HospitalSichuan UniversityChengduPR China
- Key Laboratory of Rehabilitation Medicine in Sichuan ProvinceSichuan UniversityChengduPR China
| | - Quan Wei
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China HospitalSichuan UniversityChengduPR China
- Key Laboratory of Rehabilitation Medicine in Sichuan ProvinceSichuan UniversityChengduPR China
| |
Collapse
|
7
|
Neuritin Promotes Bone Marrow-Derived Mesenchymal Stem Cell Migration to Treat Diabetic Peripheral Neuropathy. Mol Neurobiol 2022; 59:6666-6683. [DOI: 10.1007/s12035-022-03002-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 08/16/2022] [Indexed: 10/15/2022]
|
8
|
Zarepour A, Bal Öztürk A, Koyuncu Irmak D, Yaşayan G, Gökmen A, Karaöz E, Zarepour A, Zarrabi A, Mostafavi E. Combination Therapy Using Nanomaterials and Stem Cells to Treat Spinal Cord Injuries. Eur J Pharm Biopharm 2022; 177:224-240. [PMID: 35850168 DOI: 10.1016/j.ejpb.2022.07.004] [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: 05/27/2022] [Revised: 06/29/2022] [Accepted: 07/08/2022] [Indexed: 02/07/2023]
Abstract
As a part of the central nervous system, the spinal cord (SC) provides most of the communications between the brain and other parts of the body. Any damage to SC interrupts this communication, leading to serious problems, which may remain for the rest of their life. Due to its significant impact on patients' quality of life and its exorbitant medical costs, SC injury (SCI) is known as one of the most challengeable diseases in the world. Thus, it is critical to introduce highly translatable therapeutic platforms for SCI treatment. So far, different strategies have been introduced, among which utilizing various types of stem cells is one of the most interesting ones. The capability of stem cells to differentiate into several types of cell lines makes them promising candidates for the regeneration of injured tissues. One of the other interesting and novel strategies for SCI treatment is the application of nanomaterials, which could appear as a carrier for therapeutic agents or as a platform for culturing the cells. Combining these two approaches, stem cells and nanomaterials, could provide promising therapeutic strategies for SCI management. Accordingly, in this review we have summarized some of the recent advancements in which the applications of different types of stem cells and nanomaterials, alone and in combination forms, were evaluated for SCI treatment.
Collapse
Affiliation(s)
- Arezou Zarepour
- Radiology Department, Kashan University of Medical Sciences, Kashan, Isfahan, Iran
| | - Ayça Bal Öztürk
- Department of Stem Cell and Tissue Engineering, Institute of Health Sciences, Istinye University, Istanbul, Turkey; Department of Analytical Chemistry, Faculty of Pharmacy, Istinye University, Zeytinburnu, Turkey
| | | | - Gökçen Yaşayan
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Yeditepe University, Istanbul, Turkey
| | - Aylin Gökmen
- Molecular Biology and Genetics Department, Faculty of Engineering and Natural Sciences, Bahcesehir University, Besiktas, Istanbul, Turkey
| | - Erdal Karaöz
- Liv Hospital, Center for Regenerative Medicine and Stem Cell Manufacturing (LivMedCell), İstanbul, Turkey
| | - Atefeh Zarepour
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Sariyer, Istanbul 34396, Turkey
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Sariyer, Istanbul 34396, Turkey.
| | - Ebrahim Mostafavi
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
9
|
Xie JL, Wang XR, Li MM, Tao ZH, Teng WW, Saijilafu. Mesenchymal Stromal Cell Therapy in Spinal Cord Injury: Mechanisms and Prospects. Front Cell Neurosci 2022; 16:862673. [PMID: 35722621 PMCID: PMC9204037 DOI: 10.3389/fncel.2022.862673] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 05/09/2022] [Indexed: 12/13/2022] Open
Abstract
Spinal cord injury (SCI) often leads to severe motor, sensory, and autonomic dysfunction in patients and imposes a huge economic cost to individuals and society. Due to its complicated pathophysiological mechanism, there is not yet an optimal treatment available for SCI. Mesenchymal stromal cells (MSCs) are promising candidate transplant cells for use in SCI treatment. The multipotency of MSCs, as well as their rich trophic and immunomodulatory abilities through paracrine signaling, are expected to play an important role in neural repair. At the same time, the simplicity of MSCs isolation and culture and the bypassing of ethical barriers to stem cell transplantation make them more attractive. However, the MSCs concept has evolved in a specific research context to encompass different populations of cells with a variety of biological characteristics, and failure to understand this can undermine the quality of research in the field. Here, we review the development of the concept of MSCs in order to clarify misconceptions and discuss the controversy in MSCs neural differentiation. We also summarize a potential role of MSCs in SCI treatment, including their migration and trophic and immunomodulatory effects, and their ability to relieve neuropathic pain, and we also highlight directions for future research.
Collapse
Affiliation(s)
- Ji-Le Xie
- Department of Orthopaedics, The First Affiliated Hospital, Soochow University, Suzhou, China,Orthopaedic Institute, School of Medicine, Soochow University, Suzhou, China
| | - Xing-Ran Wang
- Orthopaedic Institute, School of Medicine, Soochow University, Suzhou, China
| | - Mei-Mei Li
- Orthopaedic Institute, School of Medicine, Soochow University, Suzhou, China
| | - Zi-Han Tao
- Orthopaedic Institute, School of Medicine, Soochow University, Suzhou, China
| | - Wen-Wen Teng
- Orthopaedic Institute, School of Medicine, Soochow University, Suzhou, China
| | - Saijilafu
- Department of Orthopaedics, The First Affiliated Hospital, Soochow University, Suzhou, China,Orthopaedic Institute, School of Medicine, Soochow University, Suzhou, China,*Correspondence: Saijilafu,
| |
Collapse
|
10
|
Nishimura Y, Nakamura T, Kamijo YI, Arakawa H, Umemoto Y, Kinoshita T, Sakurai Y, Tajima F. Increased serum levels of brain-derived neurotrophic factor following wheelchair half marathon race in individuals with spinal cord injury. J Spinal Cord Med 2022; 45:455-460. [PMID: 33054672 PMCID: PMC9135440 DOI: 10.1080/10790268.2020.1816402] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Objective: Brain-derived neurotrophic factor (BDNF) has beneficial effects on metabolism as well as the peripheral and central nervous systems. The aim of this study was to assess the response of serum BDNF concentration ([BDNF]s) to wheelchair half marathon race in individuals with spinal cord injury (SCI).Design: Prospective observational study.Setting: The 34th Oita International Wheelchair Marathon Race in Japan.Participants: Nine cervical SCIs (CSCI) and 8 thoracic and lumber SCIs (LSCI) male athletes. Interventions: Wheelchair half-Marathon Race.Outcome measures: [BDNF]s, plasma concentrations of adrenaline ([Ad]p), noradrenaline ([Nor]p), and cortisol ([Cor]p), hematocrit, and platelet count were measured the day before, immediately after, and an hour after the race.Results: [BDNF]s increased significantly immediately after the race in both groups (CSCI; P = 0.0055, LSCI; P = 0.0312) but returned to the baseline levels at one hour after the race. However, [BDNF]s immediately and one hour after the race were significantly higher in LSCI than in CSCI (immediately after the race; P = 0.0037, 1 h after the race; P = 0.0206). Hematocrit and platelet count remained unchanged throughout the study. In LSCI, [Ad]p, [Nor]p and [Cor]p increased significantly immediately after and one hour after the race, compared with the baseline values (P < 0.05). On the other hand, these variables remained unchanged throughout the study in the CSCI.Conclusions: [BDNF]s increased significantly from the baseline in both LCSI and CSCI but was higher in LSCI than in CSCI immediately after and one hour after the race.
Collapse
Affiliation(s)
- Yukihide Nishimura
- Department of Rehabilitation Medicine, Iwate Medical University, Iwate, Japan
| | - Takeshi Nakamura
- Department of Rehabilitation Medicine, School of Medicine, Yokohama City University, Kanagawa, Japan
| | - Yoshi-ichiro Kamijo
- Department of Rehabilitation Medicine, Wakayama Medical University, Wakayama, Japan
| | - Hideki Arakawa
- Department of Rehabilitation Medicine, School of Medicine, Yokohama City University, Kanagawa, Japan
| | - Yasunori Umemoto
- Department of Rehabilitation Medicine, Wakayama Medical University, Wakayama, Japan
| | - Tokio Kinoshita
- Department of Rehabilitation Medicine, Wakayama Medical University, Wakayama, Japan
| | - Yuta Sakurai
- Research Center of Sports Medicine and Balneology, Nachikatsuura Balneologic Town Hospital, Wakayama, Japan
| | - Fumihiro Tajima
- Department of Rehabilitation Medicine, Wakayama Medical University, Wakayama, Japan,Correspondence to: Fumihiro Tajima, Department of Rehabilitation Medicine, Wakayama Medical University, 811-1 Kimiidera Wakayama city, Wakayama641-8509, Japan; Ph: +81-73-441-0664.
| |
Collapse
|
11
|
Progression in translational research on spinal cord injury based on microenvironment imbalance. Bone Res 2022; 10:35. [PMID: 35396505 PMCID: PMC8993811 DOI: 10.1038/s41413-022-00199-9] [Citation(s) in RCA: 106] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 11/14/2021] [Accepted: 12/22/2021] [Indexed: 02/07/2023] Open
Abstract
Spinal cord injury (SCI) leads to loss of motor and sensory function below the injury level and imposes a considerable burden on patients, families, and society. Repair of the injured spinal cord has been recognized as a global medical challenge for many years. Significant progress has been made in research on the pathological mechanism of spinal cord injury. In particular, with the development of gene regulation, cell sequencing, and cell tracing technologies, in-depth explorations of the SCI microenvironment have become more feasible. However, translational studies related to repair of the injured spinal cord have not yielded significant results. This review summarizes the latest research progress on two aspects of SCI pathology: intraneuronal microenvironment imbalance and regenerative microenvironment imbalance. We also review repair strategies for the injured spinal cord based on microenvironment imbalance, including medications, cell transplantation, exosomes, tissue engineering, cell reprogramming, and rehabilitation. The current state of translational research on SCI and future directions are also discussed. The development of a combined, precise, and multitemporal strategy for repairing the injured spinal cord is a potential future direction.
Collapse
|
12
|
Haggerty AE, Maldonado-Lasunción I, Nitobe Y, Yamane K, Marlow MM, You H, Zhang C, Cho B, Li X, Reddy S, Mao HQ, Oudega M. The Effects of the Combination of Mesenchymal Stromal Cells and Nanofiber-Hydrogel Composite on Repair of the Contused Spinal Cord. Cells 2022; 11:1137. [PMID: 35406701 PMCID: PMC8997442 DOI: 10.3390/cells11071137] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/17/2022] [Accepted: 03/23/2022] [Indexed: 02/01/2023] Open
Abstract
A bone marrow-derived mesenchymal stromal cell (MSC) transplant and a bioengineered nanofiber-hydrogel composite (NHC) have been shown to stimulate nervous tissue repair in the contused spinal cord in rodent models. Here, these two modalities were combined to assess their repair effects in the contused spinal cord in adult rats. Cohorts of contused rats were treated with MSC in NHC (MSC-NHC), MSC in phosphate-buffered saline (MSC-PBS), NHC, or PBS injected into the contusion site at 3 days post-injury. One week after injury, there were significantly fewer CD68+ cells in the contusion with MSC-NHC and NHC, but not MSC-PBS. The reduction in CD86+ cells in the injury site with MSC-NHC was mainly attributed to NHC. One and eight weeks after injury, we found a greater CD206+/CD86+ cell ratio with MSC-NHC or NHC, but not MSC-PBS, indicating a shift from a pro-inflammatory towards an anti-inflammatory milieu in the injury site. Eight weeks after injury, the injury size was significantly reduced with MSC-NHC, NHC, and MSC-PBS. At this time, astrocyte, and axon presence in the injury site was greater with MSC-NHC compared with MSC-PBS. We did not find a significant effect of NHC on MSC transplant survival, and hind limb function was similar across all groups. However, we did find fewer macrophages at 1 week post-injury, more macrophages polarized towards a pro-regenerative phenotype at 1 and 8 weeks after injury, and reduced injury volume, more astrocytes, and more axons at 8 weeks after injury in rats with MSC-NHC and NHC alone compared with MSC-PBS; these findings were especially significant between rats with MSC-NHC and MSC-PBS. The data support further study in the use of an NHC-MSC combination transplant in the contused spinal cord.
Collapse
Affiliation(s)
- Agnes E. Haggerty
- The Miami Project to Cure Paralysis, University of Miami, Miami, FL 33136, USA; (A.E.H.); (I.M.-L.); (Y.N.); (K.Y.); (M.M.M.)
| | - Ines Maldonado-Lasunción
- The Miami Project to Cure Paralysis, University of Miami, Miami, FL 33136, USA; (A.E.H.); (I.M.-L.); (Y.N.); (K.Y.); (M.M.M.)
- Department of Regeneration of Sensorimotor Systems, Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences, 1105 BA Amsterdam, The Netherlands
- Shirley Ryan AbilityLab, Chicago, IL 60611, USA
- Department of Physical Therapy and Human Movements Sciences, Northwestern University, Chicago, IL 60611, USA
| | - Yohshiro Nitobe
- The Miami Project to Cure Paralysis, University of Miami, Miami, FL 33136, USA; (A.E.H.); (I.M.-L.); (Y.N.); (K.Y.); (M.M.M.)
- Department of Orthopedic Surgery, Hirosaki University Graduate School of Medicine, Hirosaki 036-8562, Japan
| | - Kentaro Yamane
- The Miami Project to Cure Paralysis, University of Miami, Miami, FL 33136, USA; (A.E.H.); (I.M.-L.); (Y.N.); (K.Y.); (M.M.M.)
- Department of Orthopedic Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Science, Kitaku, Okayama 700-8558, Japan
| | - Megan M. Marlow
- The Miami Project to Cure Paralysis, University of Miami, Miami, FL 33136, USA; (A.E.H.); (I.M.-L.); (Y.N.); (K.Y.); (M.M.M.)
| | - Hua You
- Department of Oncology and Hematology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou 510095, China;
| | - Chi Zhang
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD 21231, USA; (C.Z.); (B.C.); (X.L.)
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA;
| | - Brian Cho
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD 21231, USA; (C.Z.); (B.C.); (X.L.)
- Department of Plastic and Reconstructive Surgery, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Xiaowei Li
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD 21231, USA; (C.Z.); (B.C.); (X.L.)
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA;
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sashank Reddy
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA;
- Department of Plastic and Reconstructive Surgery, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Hai-Quan Mao
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD 21231, USA; (C.Z.); (B.C.); (X.L.)
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA;
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Martin Oudega
- Shirley Ryan AbilityLab, Chicago, IL 60611, USA
- Department of Physical Therapy and Human Movements Sciences, Northwestern University, Chicago, IL 60611, USA
- Department of Neuroscience, Northwestern University, Chicago, IL 60611, USA
- Edward Hines Jr. VA Hospital, Hines, IL 60141, USA
| |
Collapse
|
13
|
Deng WS, Yang K, Liang B, Liu YF, Chen XY, Zhang S. Collagen/heparin sulfate scaffold combined with mesenchymal stem cells treatment for canines with spinal cord injury: A pilot feasibility study. J Orthop Surg (Hong Kong) 2021; 29:23094990211012293. [PMID: 34060363 DOI: 10.1177/23094990211012293] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Due to endogenous neuronal deficiency and glial scar formation, spinal cord injury (SCI) often leads to irreversible neurological loss. Accumulating evidence has shown that a suitable scaffold has important value for promoting nerve regeneration after SCI. Collagen/heparin sulfate scaffold (CHSS) has shown effect for guiding axonal regeneration and decreasing glial scar deposition after SCI. The current research aimed to evaluate the utility of the CHSSs adsorbed with mesenchymal stem cells (MSCs) on nerve regeneration, and functional recovery after acute complete SCI. METHODS CHSSs were prepared, and evaluated for biocompatibility. The CHSSs adsorbed with MSCs were transplanted into these canines with complete SCI. RESULTS We observed that MSCs had good biocompatibility with CHSSs. In complete transverse SCI models, the implantation of CHSS co-cultured with MSCs exhibited significant improvement in locomotion, motor evoked potential, magnetic resonance imaging, diffusion tensor imaging, and urodynamic parameters. Meanwhile, nerve fibers were markedly improved in the CHSS adsorbed with MSCs group. Moreover, we observed that the implantation of CHSS combined with MSCs modulated inflammatory cytokine levels. CONCLUSIONS The results preliminarily demonstrated that the transplantation of MSCs on a CHSS could improve the recovery of motor function after SCI. Thus, implanting the MSCs-laden CHSS is a promising combinatorial therapy for treatment in acute SCI.
Collapse
Affiliation(s)
- Wu-Sheng Deng
- College of Integrated Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu Province, China
| | - Kun Yang
- The First Affiliated Hospital of Hainan Medical College, Haikou, China
| | - Bing Liang
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of Chinese People's Armed Police Force, Tianjin, China
| | - Ying-Fu Liu
- Cang Zhou Nanobody Technology Innovation Center, Cangzhou, China
| | - Xu-Yi Chen
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of Chinese People's Armed Police Force, Tianjin, China
| | - Sai Zhang
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, Characteristic Medical Center of Chinese People's Armed Police Force, Tianjin, China
| |
Collapse
|
14
|
Shinozaki M, Nagoshi N, Nakamura M, Okano H. Mechanisms of Stem Cell Therapy in Spinal Cord Injuries. Cells 2021; 10:cells10102676. [PMID: 34685655 PMCID: PMC8534136 DOI: 10.3390/cells10102676] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 09/28/2021] [Accepted: 10/04/2021] [Indexed: 12/13/2022] Open
Abstract
Every year, 0.93 million people worldwide suffer from spinal cord injury (SCI) with irretrievable sequelae. Rehabilitation, currently the only available treatment, does not restore damaged tissues; therefore, the functional recovery of patients remains limited. The pathophysiology of spinal cord injuries is heterogeneous, implying that potential therapeutic targets differ depending on the time of injury onset, the degree of injury, or the spinal level of injury. In recent years, despite a significant number of clinical trials based on various types of stem cells, these aspects of injury have not been effectively considered, resulting in difficult outcomes of trials. In a specialty such as cancerology, precision medicine based on a patient’s characteristics has brought indisputable therapeutic advances. The objective of the present review is to promote the development of precision medicine in the field of SCI. Here, we first describe the multifaceted pathophysiology of SCI, with the temporal changes after injury, the characteristics of the chronic phase, and the subtypes of complete injury. We then detail the appropriate targets and related mechanisms of the different types of stem cell therapy for each pathological condition. Finally, we highlight the great potential of stem cell therapy in cervical SCI.
Collapse
Affiliation(s)
- Munehisa Shinozaki
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan;
| | - Narihito Nagoshi
- Department of Orthopedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; (N.N.); (M.N.)
| | - Masaya Nakamura
- Department of Orthopedic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; (N.N.); (M.N.)
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan;
- Correspondence:
| |
Collapse
|
15
|
Biktimirov A, Pak O, Bryukhovetskiy I, Sharma A, Sharma HS. Neuromodulation as a basic platform for neuroprotection and repair after spinal cord injury. PROGRESS IN BRAIN RESEARCH 2021; 266:269-300. [PMID: 34689861 DOI: 10.1016/bs.pbr.2021.06.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Spinal cord injury (SCI) is one of the most challenging medical issues. Spasticity is a major complication of SCI. A combination of spinal cord stimulation, new methods of neuroprotection and biomedical cellular products provides fundamentally new options for SCI treatment and rehabilitation. The paper attempts to critically analyze the effectiveness of using these procedures for patients with SCI, suggesting a protocol for a step-by-step personalized treatment of SCI, based on continuity of modern conservative and surgical methods. The study argues the possibility of using neuromodulation as a basis for rehabilitating patients with SCI.
Collapse
Affiliation(s)
- Artur Biktimirov
- Department of Fundamental Medicine, School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia.
| | - Oleg Pak
- Department of Neurosurgery, Medical Center, Far Eastern Federal University, Vladivostok, Russia
| | - Igor Bryukhovetskiy
- Department of Fundamental Medicine, School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
| | - Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
16
|
Ebhodaghe SO. Natural Polymeric Scaffolds for Tissue Engineering Applications. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2021; 32:2144-2194. [PMID: 34328068 DOI: 10.1080/09205063.2021.1958185] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Natural polymeric scaffolds can be used for tissue engineering applications such as cell delivery and cell-free supporting of native tissues. This is because of their desirable properties such as; high biocompatibility, tunable mechanical strength and conductivity, large surface area, porous- and extracellular matrix (ECM)-mimicked structures. Specifically, their less toxicity and biocompatibility makes them suitable for several tissue engineering applications. For these reasons, several biopolymeric scaffolds are currently being explored for numerous tissue engineering applications. To date, research on the nature, chemistry, and properties of nanocomposite biopolymers are been reported, while the need for a comprehensive research note on more tissue engineering application of these biopolymers remains. As a result, this present study comprehensively reviews the development of common natural biopolymers as scaffolds for tissue engineering applications such as cartilage tissue engineering, cornea repairs, osteochondral defect repairs, and nerve regeneration. More so, the implications of research findings for further studies are presented, while the impact of research advances on future research and other specific recommendations are added as well.
Collapse
|
17
|
Li WY, Jia H, Wang ZD, Zhai FG, Sun GD, Ma D, Liu GB, Li CM, Wang Y. Combinatory transplantation of mesenchymal stem cells with flavonoid small molecule in acellular nerve graft promotes sciatic nerve regeneration. J Tissue Eng 2020; 11:2041731420980136. [PMID: 34956585 PMCID: PMC8693221 DOI: 10.1177/2041731420980136] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 11/21/2020] [Indexed: 12/11/2022] Open
Abstract
Previous animal studies have demonstrated that the flavonoid small-molecule TrkB agonist, 7, 8-dihydroxyflavone (DHF), promotes axon regeneration in transected peripheral nerves. In the present study, we investigated the combined effects of 7, 8-DHF treatment and bone marrow-derived stem/stromal cells (BMSCs) engraftment into acellular nerve allografts (ANAs) and explore relevant mechanisms that may be involved. Our results show that TrkB and downstream ERK1/2 phosphorylation are increased upon 7, 8-DHF treatment compared to the negative control group. Also, 7, 8-DHF promotes proliferation, survival, and Schwann-like cell differentiation of BMSCs in vitro. While selective ERK1/2 inhibitor U0126 suppressed the effect of upregulation of ERK1/2 phosphorylation and decreased cell proliferation, survival, and Schwann-like cell differentiation partially induced by 7, 8-DHF. In vivo, 7, 8-DHF promotes survival of transplanted BMSCs and upregulates axonal growth and myelination in regenerating ANAs. 7, 8-DHF+BMSCs also improved motor endplate density of target musculature. These benefits were associated with increased motor functional recovery. 7, 8-DHF+BMSCs significantly upregulated TrkB and ERK1/2 phosphorylation expression in regenerating ANA, and increased TrkB expression in the lumbar spinal cord. The mechanism of 7, 8-DHF action may be related to its ability to upregulate TrkB signaling, and downstream activation of survival signaling molecules ERK1/2 in the regenerating ANAs and spinal cord and improved survival of transplanted BMSCs. This study provides novel foundational data connecting the benefits of 7, 8-DHF treatment in neural injury and repair to BMSCs biology and function and demonstrates a potential combination approach for the treatment of injured peripheral nerve via nerve graft transplant.
Collapse
Affiliation(s)
- Wen-yuan Li
- Institute of Neural Tissue Engineering, Mudanjiang College of Medicine, Mudanjiang, China
| | - Hua Jia
- Department of Anatomy, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
- Center for Reproductive Biology and Health, College of Agricultural Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Zhen-Dong Wang
- Department of Otorhinolaryngology, The Second Affiliated Hospital, Mudanjiang College of Medicine, Mudanjiang, China
| | - Feng-guo Zhai
- Department of Pharmacology, Mudanjiang College of Medicine, Mudanjiang, China
| | - Guang-da Sun
- Institute of Neural Tissue Engineering, Mudanjiang College of Medicine, Mudanjiang, China
| | - Duo Ma
- Institute of Neural Tissue Engineering, Mudanjiang College of Medicine, Mudanjiang, China
| | - Gui-Bo Liu
- Institute of Neural Tissue Engineering, Mudanjiang College of Medicine, Mudanjiang, China
| | - Chun-Mei Li
- Department of Basic Psychological, Mudanjiang College of Medicine, Mudanjiang, China
| | - Ying Wang
- Institute of Neural Tissue Engineering, Mudanjiang College of Medicine, Mudanjiang, China
| |
Collapse
|
18
|
Huang F, Chen T, Chang J, Zhang C, Liao F, Wu L, Wang W, Yin Z. A conductive dual-network hydrogel composed of oxidized dextran and hyaluronic-hydrazide as BDNF delivery systems for potential spinal cord injury repair. Int J Biol Macromol 2020; 167:434-445. [PMID: 33278434 DOI: 10.1016/j.ijbiomac.2020.11.206] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 11/27/2020] [Accepted: 11/29/2020] [Indexed: 12/17/2022]
Abstract
Spinal cord injury (SCI) often causes neuronal death and axonal degeneration. In this study, we report a new strategy for preparing injectable and conductive polysaccharides-based hydrogels that could sustainably deliver brain-derived neurotrophic factor (BDNF) for SCI repair. We used poly(lactic-co-glycolic acid) (PLGA) as a carrier to encapsulate BDNF. The resulting microspheres were then modified with tannic acid (TA). The polysaccharides-based hydrogel composed of oxidized dextran (Dex) and hyaluronic acid-hydrazide (HA) was mixed with TA-modified microspheres to form the ultimate BDNF@TA-PLGA/Dex-HA hydrogel. Our results showed that the hydrogel had properties similar to natural spinal cords. Specifically, the hydrogel had soft mechanical properties and high electrical conductivity. The cross-sectional morphology of the hydrogel exhibited a continuous and porous structure. The swelling and degradation behaviors of the Dex-HA hydrogel in vitro indicated the incorporation of TA into hydrogel matrix could improve the stability of the hydrogel matrix as well as extend the release time of BDNF from the matrix. Furthermore, results from immunostaining and real-time PCR demonstrated that BDNF@TA-PLGA/Dex-HA hydrogel could promote the differentiation of neural stem cells (NSCs) into neurons and inhibit astrocyte differentiation in vitro. These results show the great potential of this hydrogel as a biomimetic material in SCI regeneration.
Collapse
Affiliation(s)
- Fei Huang
- Department of Orthopaedics, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Taiying Chen
- Department of Liver Transplantation, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Jun Chang
- Department of Orthopaedics, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Chi Zhang
- Department of Orthopaedics, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Faxue Liao
- Department of Orthopaedics, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Linwei Wu
- Department of Liver Transplantation, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China.
| | - Wenbin Wang
- Department of General Surgery, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
| | - Zongsheng Yin
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
19
|
Bartlett RD, Burley S, Ip M, Phillips JB, Choi D. Cell Therapies for Spinal Cord Injury: Trends and Challenges of Current Clinical Trials. Neurosurgery 2020; 87:E456-E472. [DOI: 10.1093/neuros/nyaa149] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Accepted: 03/11/2020] [Indexed: 12/22/2022] Open
Abstract
Abstract
Cell therapies have the potential to revolutionize the treatment of spinal cord injury. Basic research has progressed significantly in recent years, with a plethora of cell types now reaching early-phase human clinical trials, offering new strategies to repair the spinal cord. However, despite initial enthusiasm for preclinical and early-phase clinical trials, there has been a notable hiatus in the translation of cell therapies to routine clinical practice. Here, we review cell therapies that have reached clinical trials for spinal cord injury, providing a snapshot of all registered human trials and a summary of all published studies. Of registered trials, the majority have used autologous cells and approximately a third have been government funded, a third industry sponsored, and a third funded by university or healthcare systems. A total of 37 cell therapy trials have been published, primarily using stem cells, although a smaller number have used Schwann cells or olfactory ensheathing cells. Significant challenges remain for cell therapy trials in this area, including achieving stringent regulatory standards, ensuring appropriately powered efficacy trials, and establishing sustainable long-term funding. However, cell therapies hold great promise for human spinal cord repair and future trials must continue to capitalize on the exciting developments emerging from preclinical studies.
Collapse
Affiliation(s)
- Richard D Bartlett
- Centre for Nerve Engineering, University College London, London, United Kingdom
- Department of Pharmacology, UCL School of Pharmacy, University College London, London, United Kingdom
- Brain Repair and Rehabilitation, Institute of Neurology, University College London, London, United Kingdom
| | - Sarah Burley
- Centre for Nerve Engineering, University College London, London, United Kingdom
| | - Mina Ip
- Centre for Nerve Engineering, University College London, London, United Kingdom
| | - James B Phillips
- Centre for Nerve Engineering, University College London, London, United Kingdom
- Department of Pharmacology, UCL School of Pharmacy, University College London, London, United Kingdom
| | - David Choi
- Centre for Nerve Engineering, University College London, London, United Kingdom
- Brain Repair and Rehabilitation, Institute of Neurology, University College London, London, United Kingdom
- Victor Horsley Department of Neurosurgery, National Hospital for Neurology and Neurosurgery, London, United Kingdom
| |
Collapse
|
20
|
Zhan J, Li X, Luo D, Hou Y, Hou Y, Chen S, Xiao Z, Luan J, Lin D. Polydatin promotes the neuronal differentiation of bone marrow mesenchymal stem cells in vitro and in vivo: Involvement of Nrf2 signalling pathway. J Cell Mol Med 2020; 24:5317-5329. [PMID: 32299154 PMCID: PMC7205798 DOI: 10.1111/jcmm.15187] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 01/26/2020] [Accepted: 02/23/2020] [Indexed: 12/11/2022] Open
Abstract
Bone marrow mesenchymal stem cell (BMSC) transplantation represents a promising repair strategy following spinal cord injury (SCI), although the therapeutic effects are minimal due to their limited neural differentiation potential. Polydatin (PD), a key component of the Chinese herb Polygonum cuspidatum, exerts significant neuroprotective effects in various central nervous system disorders and protects BMSCs against oxidative injury. However, the effect of PD on the neuronal differentiation of BMSCs, and the underlying mechanisms remain inadequately understood. In this study, we induced neuronal differentiation of BMSCs in the presence of PD, and analysed the Nrf2 signalling and neuronal differentiation markers using routine molecular assays. We also established an in vivo model of SCI and assessed the locomotor function of the mice through hindlimb movements and electrophysiological measurements. Finally, tissue regeneration was evaluated by H&E staining, Nissl staining and transmission electron microscopy. PD (30 μmol/L) markedly facilitated BMSC differentiation into neuron‐like cells by activating the Nrf2 pathway and increased the expression of neuronal markers in the transplanted BMSCs at the injured spinal cord sites. Furthermore, compared with either monotherapy, the combination of PD and BMSC transplantation promoted axonal rehabilitation, attenuated glial scar formation and promoted axonal generation across the glial scar, thereby enhancing recovery of hindlimb locomotor function. Taken together, PD augments the neuronal differentiation of BMSCs via Nrf2 activation and improves functional recovery, indicating a promising new therapeutic approach against SCI.
Collapse
Affiliation(s)
- Jiheng Zhan
- Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xing Li
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Spine Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Dan Luo
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Spine Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yu Hou
- Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Spine Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yonghui Hou
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Spine Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shudong Chen
- Department of Spine Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhifeng Xiao
- Department of Spine Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiyao Luan
- Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Dingkun Lin
- Department of Spine Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
21
|
Ji WC, Li M, Jiang WT, Ma X, Li J. Protective effect of brain-derived neurotrophic factor and neurotrophin-3 overexpression by adipose-derived stem cells combined with silk fibroin/chitosan scaffold in spinal cord injury. Neurol Res 2020; 42:361-371. [PMID: 32149594 DOI: 10.1080/01616412.2020.1735819] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Objectives: Spinal cord injury (SCI) is a most debilitating traumatic injury, and cytotherapy is a promising alternative treatment strategy. Here we investigated the effect and mechanism of adipose-derived stem/stromal cells (ASCs) with overexpressing brain-derived neurotrophic factor (BDNF) and neurotrophin-3 (NT3) (BDNF-NT3) in combination with silk fibroin/chitosan scaffold (SFCS) in SCI.Methods: Female Sprague-Dawley rats were used as an SCI model. SFCS,SFCS and ASCs, or ASCs overexpressing NT3, BDNF, and BDNF-NT3 were implanted into SCI rats. Basso, Beattie, and Bresnahan score, pathological changes, and spinal cord tissue and nerve fiber morphology were observed and assayed. GAP-43, GFAP, and caspase-3 expression was determined using immunohistochemistry and western blotting.Results: Smoother spinal cords, less scar tissue, and lower inflammatory activity were found in the SFCS, SFCS and ASCs, ASCs with NT3, BDNF, and BDNF-NT3 overexpression treatment than in the untreated SCI rat groups. Increasing formation of nerve fibers was observed in the above groups in order. GAP-43 expression significantly increased, while GFAP and caspase-3 expression significantly decreased. These results indicated obvious alleviation in pathological changes and BDNF-NT3 overexpression in ASCs combined with SFCS treatment in SCI rats.Conclusion: Thus, BDNF-NT3 overexpression from ASCs with SFCS had synergistic neuroprotective effects on SCI and may be a treatment option for SCI.
Collapse
Affiliation(s)
- Wen-Chen Ji
- Department of Orthopaedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Meng Li
- Department of Orthopaedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Wan-Ting Jiang
- Department of Ultrasound Diagnosis, The Fourth Hospital of Xi'an, Xi'an, China
| | - Xing Ma
- Department of Orthopaedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jia Li
- Department of Orthopaedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
22
|
Chen X, Wu J, Sun R, Zhao Y, Li Y, Pan J, Chen Y, Wang X. Tubular scaffold with microchannels and an H-shaped lumen loaded with bone marrow stromal cells promotes neuroregeneration and inhibits apoptosis after spinal cord injury. J Tissue Eng Regen Med 2020; 14:397-411. [PMID: 31821733 PMCID: PMC7155140 DOI: 10.1002/term.2996] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 11/13/2019] [Accepted: 11/19/2019] [Indexed: 12/11/2022]
Abstract
As a result of its complex histological structure, regeneration patterns of grey and white matter are quite different in the spinal cord. Therefore, tissue engineering scaffolds for repairing spinal cord injury must be able to adapt to varying neural regeneration patterns. The aim of the present study was to improve a previously reported spinal cord‐mimicking partition‐type scaffold by adding microchannels on a single tubular wall along its longitudinal axis, thus integrating the two architectures of a single H‐shaped central tube and many microchannels. Next, the integrated scaffold was loaded with bone marrow stromal cells (BMSCs) and transplanted to bridge the 5‐mm defect of a complete transverse lesion in the thoracic spinal cord of rats. Subsequently, effects on nerve regeneration, locomotion function recovery, and early neuroprotection were observed. After 1 year of repair, the integrated scaffold could guide the regeneration of axons appearing in the debris of degraded microchannels, especially serotonin receptor 1A receptor‐positive axonal tracts, which were relatively orderly arranged. Moreover, a network of nerve fibres was present, and a few BMSCs expressed neuronal markers in tubular lumens. Functionally, electrophysiological and locomotor functions of rats were partially recovered. In addition, we found that BMSCs could protect neurons and oligodendrocytes from apoptosis during the early stage of implantation. Taken together, our results demonstrate the potential of this novel integrated scaffold loaded with BMSCs to promote spinal cord regeneration through mechanical guidance and neuroprotective mechanisms.
Collapse
Affiliation(s)
- Xue Chen
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu, China.,Wuxi Medical College, Jiangnan University, Wuxi, Jiangsu, China
| | - Jian Wu
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu, China
| | - Rongcheng Sun
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu, China
| | - Yahong Zhao
- Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, Jiangsu, China
| | - Yi Li
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu, China
| | - Jingying Pan
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu, China
| | - Ying Chen
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu, China
| | - Xiaodong Wang
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu, China.,Key Laboratory for Neuroregeneration of Ministry of Education and Co-innovation Center for Neuroregeneration of Jiangsu Province, Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
23
|
Deng WS, Ma K, Liang B, Liu XY, Xu HY, Zhang J, Shi HY, Sun HT, Chen XY, Zhang S. Collagen scaffold combined with human umbilical cord-mesenchymal stem cells transplantation for acute complete spinal cord injury. Neural Regen Res 2020; 15:1686-1700. [PMID: 32209773 PMCID: PMC7437585 DOI: 10.4103/1673-5374.276340] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Currently, there is no effective strategy to promote functional recovery after a spinal cord injury. Collagen scaffolds can not only provide support and guidance for axonal regeneration, but can also serve as a bridge for nerve regeneration at the injury site. They can additionally be used as carriers to retain mesenchymal stem cells at the injury site to enhance their effectiveness. Hence, we hypothesized that transplanting human umbilical cord-mesenchymal stem cells on collagen scaffolds would enhance healing following acute complete spinal cord injury. Here, we test this hypothesis through animal studies and a phase I clinical trial. (1) Animal experiments: Models of completely transected spinal cord injury were established in rats and canines by microsurgery. Mesenchymal stem cells derived from neonatal umbilical cord tissue were adsorbed onto collagen scaffolds and surgically implanted at the injury site in rats and canines; the animals were observed after 1 week–6 months. The transplantation resulted in increased motor scores, enhanced amplitude and shortened latency of the motor evoked potential, and reduced injury area as measured by magnetic resonance imaging. (2) Phase I clinical trial: Forty patients with acute complete cervical injuries were enrolled at the Characteristic Medical Center of Chinese People’s Armed Police Force and divided into two groups. The treatment group (n = 20) received collagen scaffolds loaded with mesenchymal stem cells derived from neonatal umbilical cord tissues; the control group (n = 20) did not receive the stem-cell loaded collagen implant. All patients were followed for 12 months. In the treatment group, the American Spinal Injury Association scores and activities of daily life scores were increased, bowel and urinary functions were recovered, and residual urine volume was reduced compared with the pre-treatment baseline. Furthermore, magnetic resonance imaging showed that new nerve fiber connections were formed, and diffusion tensor imaging showed that electrophysiological activity was recovered after the treatment. No serious complication was observed during follow-up. In contrast, the neurological functions of the patients in the control group were not improved over the follow-up period. The above data preliminarily demonstrate that the transplantation of human umbilical cord-mesenchymal stem cells on a collagen scaffold can promote the recovery of neurological function after acute spinal cord injury. In the future, these results need to be confirmed in a multicenter, randomized controlled clinical trial with a larger sample size. The clinical trial was approved by the Ethics Committee of the Characteristic Medical Center of Chinese People’s Armed Police Force on February 3, 2016 (approval No. PJHEC-2016-A8). All animal experiments were approved by the Ethics Committee of the Characteristic Medical Center of Chinese People’s Armed Police Force on May 20, 2015 (approval No. PJHEC-2015-D5).
Collapse
Affiliation(s)
- Wu-Sheng Deng
- College of Integrated Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu Province, China
| | - Ke Ma
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, characteristic medical center of Chinese people's armed police force, Tianjin, China
| | - Bing Liang
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, characteristic medical center of Chinese people's armed police force, Tianjin, China
| | - Xiao-Yin Liu
- Clinical School of Medicine, Tianjin Medical University, Tianjin, China
| | - Hui-You Xu
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, characteristic medical center of Chinese people's armed police force, Tianjin, China
| | - Jian Zhang
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, characteristic medical center of Chinese people's armed police force, Tianjin, China
| | - Heng-Yuan Shi
- Clinical School of Medicine, Logistics University of People's Armed Police Force, Tianjin, China
| | - Hong-Tao Sun
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, characteristic medical center of Chinese people's armed police force, Tianjin, China
| | - Xu-Yi Chen
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, characteristic medical center of Chinese people's armed police force, Tianjin, China
| | - Sai Zhang
- Tianjin Key Laboratory of Neurotrauma Repair, Pingjin Hospital Brain Center, characteristic medical center of Chinese people's armed police force, Tianjin, China
| |
Collapse
|
24
|
Yu T, Zhao C, Hou S, Zhou W, Wang B, Chen Y. Exosomes secreted from miRNA-29b-modified mesenchymal stem cells repaired spinal cord injury in rats. ACTA ACUST UNITED AC 2019; 52:e8735. [PMID: 31826179 PMCID: PMC6903804 DOI: 10.1590/1414-431x20198735] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 10/14/2019] [Indexed: 12/14/2022]
Abstract
Exosomes, a kind of extracellular vesicle, are promising therapeutic agents for spinal cord injury (SCI). This article aimed to investigate effects of exosomes secreted from miRNA-29b-modified bone marrow mesenchymal stem cells (BMSCs) on SCI. Exosomes were extracted from BMSCs transfected with miRNA-29b or negative control (miR NC). SCI rats were injected intravenously with exosomes (control exosomes, miRNA-29b exosomes) and BMSCs (miR NC, miRNA-29b) through the tail vein. The expression of miRNA-29b in spinal cord tissues of SCI rats was detected by qRT-PCR. The hind limb motor function was evaluated by Basso Beattie Bresnahan (BBB) score. The histopathological damage and neuronal regeneration in spinal cord tissues was observed by HE staining and immunohistochemistry, respectively. The injection of miRNA-29b exosomes and miRNA-29b BMSCs both significantly increased the expression of miRNA-29b in spinal cord tissues of SCI rats (P<0.05). Compared with SCI rats, rats in the miRNA-29b exosomes and the miRNA-29b groups exhibited improved SCI, including increased BBB score, NF200 and GAP-43 positive neurons, as well as decreased contractile nerve cell numbers and GFAP positive neurons (all P<0.05). The relieving degree of SCI was significantly higher in the miRNA-29b exosomes group than in the miRNA-29b BMSCs group (P<0.05). Exosomes secreted from miRNA-29b-modified BMSCs were effective in the repair of SCI in rats.
Collapse
Affiliation(s)
- Tao Yu
- Department of Spinal Surgery, Qilu Hospitial of ShanDong University, Jinan, Shandong, China.,Department of Orthopedics, Liaocheng People's Hospitial, Liaocheng, Shandong, China
| | - Cunju Zhao
- Department of Spinal Surgery, Qilu Hospitial of ShanDong University, Jinan, Shandong, China.,Department of Orthopedics, Liaocheng People's Hospitial, Liaocheng, Shandong, China
| | - Shouzhi Hou
- Department of Radiology, Liaocheng People's Hospitial, Liaocheng, Shandong, China
| | - Weijie Zhou
- Department of Orthopedics, Liaocheng People's Hospitial, Liaocheng, Shandong, China
| | - Baoxin Wang
- Department of Orthopedics, Liaocheng People's Hospitial, Liaocheng, Shandong, China
| | - Yunzhen Chen
- Department of Spinal Surgery, Qilu Hospitial of ShanDong University, Jinan, Shandong, China
| |
Collapse
|
25
|
Nitobe Y, Nagaoki T, Kumagai G, Sasaki A, Liu X, Fujita T, Fukutoku T, Wada K, Tanaka T, Kudo H, Asari T, Furukawa KI, Ishibashi Y. Neurotrophic Factor Secretion and Neural Differentiation Potential of Multilineage-differentiating Stress-enduring (Muse) Cells Derived from Mouse Adipose Tissue. Cell Transplant 2019; 28:1132-1139. [PMID: 31304790 PMCID: PMC6767880 DOI: 10.1177/0963689719863809] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Multilineage-differentiating stress-enduring (Muse) cells are endogenous pluripotent stem cells that can be isolated based on stage-specific embryonic antigen-3 (SSEA-3), a pluripotent stem cell-surface marker. However, their capacities for survival, neurotrophic factor secretion, and neuronal and glial differentiation are unclear in rodents. Here we analyzed mouse adipose tissue-derived Muse cells in vitro. We collected mesenchymal stem cells (MSCs) from C57BL/6 J mouse adipose tissue and separated SSEA-3+, namely Muse cells, and SSEA-3-, non-Muse cells, to assess self-renewability; pluripotency marker expression (Nanog, Oct3/4, Sox2, and SSEA-3); spontaneous differentiation into endodermal, mesodermal, and ectodermal lineages; and neural differentiation capabilities under cytokine induction. Neurally differentiated Muse and non-Muse cell functions were assessed by calcium imaging. Antioxidant ability was measured to assess survival under oxidative stress. Brain-derived neurotrophic factor (BDNF), vascular endothelial cell growth factor (VEGF), and hepatocyte growth factor (HGF) secretion were analyzed in enzyme-linked immunosorbent assays. SSEA-3+ Muse cells (6.3 ± 1.9% of mouse adipose-MSCs), but not non-Muse cells, exhibited self-renewability, spontaneous differentiation into the three germ layers, and differentiation into cells positive for Tuj-1 (27 ± 0.9%), O4 (17 ± 3.4%), or GFAP (23 ± 1.3%) under cytokine induction. Neurally differentiated Muse cells responded to KCl depolarization with greater increases in cytoplasmic Ca2+ levels than non-Muse cells. Cell survival under oxidative stress was significantly higher in Muse cells (50 ± 2.7%) versus non-Muse cells (22 ± 2.8%). Muse cells secreted significantly more BDNF, VEGF, and HGF (273 ± 12, 1479 ± 7.5, and 6591 ± 1216 pg/mL, respectively) than non-Muse cells (133 ± 4.0, 1165 ± 20, and 2383 ± 540 pg/mL, respectively). Mouse Muse cells were isolated and characterized for the first time. Muse cells showed greater pluripotency-like characteristics, survival, neurotrophic factor secretion, and neuronal and glial-differentiation capacities than non-Muse cells, indicating that they may have better neural-regeneration potential.
Collapse
Affiliation(s)
- Yohshiro Nitobe
- Department of Orthopaedic Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan.,*Both the authors contributed equally to this article
| | - Toshihide Nagaoki
- Department of Orthopaedic Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan.,*Both the authors contributed equally to this article
| | - Gentaro Kumagai
- Department of Orthopaedic Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Ayako Sasaki
- Department of Orthopaedic Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Xizhe Liu
- Department of Orthopaedic Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Taku Fujita
- Department of Orthopaedic Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Tatsuhiro Fukutoku
- Department of Orthopaedic Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Kanichiro Wada
- Department of Orthopaedic Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Toshihiro Tanaka
- Department of Orthopaedic Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Hitoshi Kudo
- Department of Orthopaedic Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Toru Asari
- Department of Orthopaedic Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Ken-Ichi Furukawa
- Department of Pharmacology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Yasuyuki Ishibashi
- Department of Orthopaedic Surgery, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| |
Collapse
|
26
|
Jia H, Wang Y, Chen J, Li JP, Han HQ, Tong XJ, He ZY, Ma WZ. Combination of BMSCs-laden acellular nerve xenografts transplantation and G-CSF administration promotes sciatic nerve regeneration. Synapse 2019; 73:e22093. [PMID: 30761618 DOI: 10.1002/syn.22093] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 02/03/2019] [Accepted: 02/11/2019] [Indexed: 12/12/2022]
Abstract
Peripheral nerve gaps often lead to interrupted innervation, manifesting as severe sensory and motor dysfunctions. The repairs of the nerve injuries have not achieved satisfactory curative effects in clinic. The transplantation of bone marrow stromal cells (BMSCs)-laden acellular nerve xenografts (ANX) has been proven more effective than the acellular nerve allografting. Besides, granulocyte colony-stimulating factor (G-CSF) can inhibit inflammation and apoptosis, and thus is conducive to the microenvironmental improvement of axonal regeneration. This study aims to investigate the joint effect of BMSCs-seeded ANX grafting and G-CSF administration, and explore the relevant mechanisms. Adult SD rats were divided into five groups randomly: ANX group, ANX combined with G-CSF group, BMSCs-laden ANX group, BMSCs-laden ANX combined with G-CSF group, and autograft group. Eight weeks after transplantation, the detection of praxiology and neuroelectrophysiology was conducted, and then the morphology of the regenerated nerves was analyzed. The inflammatory response and apoptosis in the nerve grafts as well as the expression of the growth-promoting factors in the regenerated tissues were further assayed. G-CSF intervention and BMSCs implanting synergistically promoted peripheral nerve regeneration and functional recovery following ANX bridging, and the restoration effect was matchable with that of the autologous nerve grafting. Moreover, local inflammation was alleviated, the apoptosis of the seeded BMSCs was decreased, and the levels of the neuromodulatory factors were elevated. In conclusion, the union application of BMSCs-implanted ANX and G-CSF ameliorated the niche of neurotization and advanced nerve regeneration substantially. The strategy achieved the favorable effectiveness as an alternative to the autotransplantation.
Collapse
Affiliation(s)
- Hua Jia
- Department of Human Anatomy and Histoembryology, College of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Ying Wang
- Research Institute of Neural Tissue Engineering, Department of Anatomy, Mudanjiang College of Medicine, Mudanjiang, China
| | - Jiao Chen
- Department of Human Anatomy and Histoembryology, College of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Jun-Ping Li
- Department of Human Anatomy and Histoembryology, College of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Huai-Qin Han
- Department of Human Anatomy and Histoembryology, College of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Xiao-Jie Tong
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Zhong-Yi He
- Department of Human Anatomy and Histoembryology, College of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Wen-Zhi Ma
- Department of Human Anatomy and Histoembryology, College of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China.,Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Key Laboratory of Reproduction and Genetics of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, China
| |
Collapse
|
27
|
He Z, Zang H, Zhu L, Huang K, Yi T, Zhang S, Cheng S. An anti-inflammatory peptide and brain-derived neurotrophic factor-modified hyaluronan-methylcellulose hydrogel promotes nerve regeneration in rats with spinal cord injury. Int J Nanomedicine 2019; 14:721-732. [PMID: 30705588 PMCID: PMC6342221 DOI: 10.2147/ijn.s187854] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background Traumatic spinal cord injury (SCI) causes neuronal death, demyelination, axonal degeneration, inflammation, glial scar formation, and cystic cavitation resulting in interruption of neural signaling and loss of nerve function. Multifactorial targeted therapy is a promising strategy for SCI. Methods The anti-inflammatory peptide KAFAKLAARLYRKALARQLGVAA (KAFAK) and brain-derived neurotrophic factor (BDNF)-modified hyaluronan-methylcellulose (HAMC) hydrogel was designed for minimally invasive, localized, and sustained intrathecal protein delivery. The physical and biological characteristics of HAMC-KAFAK/BDNF hydrogel were measured in vitro. SCI model was performed in rats and HAMC-KAFAK/BDNF hydrogel was injected into the injured site of spinal cord. The neuronal regeneration effect was evaluated by inflammatory cytokine levels, behavioral test and histological analysis at 8 weeks post operation. Results HAMC-KAFAK/BDNF hydrogel showed minimally swelling property and sustained release of the KAFAK and BDNF. HAMC-KAFAK/BDNF hydrogel significantly improved the proliferation of PC12 cells in vitro without cytotoxicity. Significant recovery in both neurological function and nerve tissue morphology in SCI rats were observed in HAMC-KAFAK/BDNF group. HAMC-KAFAK/BDNF group showed significant reduction in proinflammatory cytokines expression and cystic cavitation, decreased glial scar formation, and improved neuronal survival in the rat SCI model compared to HAMC group and SCI group. Conclusion The HAMC-KAFAK/BDNF hydrogel promotes functional recovery of rats with spinal cord injury by regulating inflammatory cytokine levels and improving axonal regeneration.
Collapse
Affiliation(s)
- Zhijiang He
- Logistics University of Chinese People's Armed Police Force (PAP), Tianjin 300309, China
| | - Hongxin Zang
- Department of Nursing, Characteristic Medical Center of Chinese People's Armed Police Force (PAP), Tianjin 300162, China
| | - Lei Zhu
- Department of Orthopaedics Characteristic Medical Center of Chinese People's Armed Police Force (PAP), Tianjin 300162, China
| | - Kui Huang
- Logistics University of Chinese People's Armed Police Force (PAP), Tianjin 300309, China
| | - Tailong Yi
- Institute of TBI and Neuroscience, Characteristic Medical Center of Chinese People's Armed Police Force (PAP), Tianjin Key Laboratory of Neurotrauma Repair, Tianjin 300162, China, ;
| | - Sai Zhang
- Institute of TBI and Neuroscience, Characteristic Medical Center of Chinese People's Armed Police Force (PAP), Tianjin Key Laboratory of Neurotrauma Repair, Tianjin 300162, China, ;
| | - Shixiang Cheng
- Institute of TBI and Neuroscience, Characteristic Medical Center of Chinese People's Armed Police Force (PAP), Tianjin Key Laboratory of Neurotrauma Repair, Tianjin 300162, China, ;
| |
Collapse
|
28
|
Kim YM, Jin JJ, Lee SJ, Seo TB, Ji ES. Treadmill exercise with bone marrow stromal cells transplantation facilitates neuroprotective effect through BDNF-ERK1/2 pathway in spinal cord injury rats. J Exerc Rehabil 2018; 14:335-340. [PMID: 30018915 PMCID: PMC6028222 DOI: 10.12965/jer.1836264.132] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 05/17/2018] [Indexed: 11/22/2022] Open
Abstract
Transplantation of bone marrow stromal cells (BMSCs) has been known as one of the effective therapeutic methods for functional recovery of spinal cord injury (SCI). Treadmill exercise also facilitates the functional recovery of SCI. Previously, we reported that combination of BMSCs transplantation with treadmill exercise potentiated the locomotor function in SCI rats. In the present study, we investigated whether recovery effect of BMSCs transplantation or treadmill exercise appears through the brain-derived neurotrophic factor (BDNF)-extracellular signal–regulated kinases 1/2 (ERK1/2) pathway. The spinal cord contusion injury was performed at the T9–T10 level using the impactor. Cultured BMSCs were transplanted directly into the lesion 1 week after SCI. Treadmill exercise was performed 6 days per a week for 6 weeks. Western blot for Bax, Bcl-2, BDNF, tyrosine kinase B (TrkB), and phosphorylated ERK1/2 (p-ERK1/2), phosphorylated JNK was performed. In the present results, combination of BMSCs transplantation with tread-mill exercise potently decreased Bax expression, potently increased Bcl-2 expression, and potently enhanced BDNF and TrkB expressions in the injured spinal cord. Combination of BMSCs transplantation with treadmill exercise further facilitated p-ERK1/2 and p-c-Jun expression levels. The present findings demonstrated the synergistic effect of treadmill exercise on neuroregenerative effect of BMSCs transplantation appeared through the activation of BDNF-ERK1/2 pathway in SCI.
Collapse
Affiliation(s)
- You-Mi Kim
- Sports Science Research Institution, Korea National Sport University, Seoul, Korea
| | - Jun-Jang Jin
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Sam-Jun Lee
- Department of Physical Education, College of Health, Welfare, and Education, Tong Myong University, Busan, Korea
| | - Tae-Beom Seo
- Department of Kinesiology, College of Natural Science, Jeju National University, Jeju, Korea
| | - Eun-Sang Ji
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, Korea
| |
Collapse
|
29
|
Khan IU, Yoon Y, Kim A, Jo KR, Choi KU, Jung T, Kim N, Son Y, Kim WH, Kweon OK. Improved Healing after the Co-Transplantation of HO-1 and BDNF Overexpressed Mesenchymal Stem Cells in the Subacute Spinal Cord Injury of Dogs. Cell Transplant 2018; 27:1140-1153. [PMID: 29909686 PMCID: PMC6158544 DOI: 10.1177/0963689718779766] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Abundant expression of proinflammatory cytokines after a spinal cord injury (SCI) creates an inhibitory microenvironment for neuroregeneration. The mesenchymal stem cells help to mitigate the inflammation and improve neural growth and survival. For this purpose, we potentiated the function of adipose-derived mesenchymal stem cells (Ad-MSCs) by transfecting them with brain-derived neurotrophic factor (BDNF) and heme oxygenase-1 (HO-1), through a lentivirus, to produce BDNF overexpressed Ad-MSCs (BDNF-MSCs), and HO-1 overexpressed Ad-MSCs (HO-1-MSCs). Sixteen SCI beagle dogs were randomly assigned into four treatment groups. We injected both HO-1 and BDNF-overexpressed MSCs as a combination group, to selectively control inflammation and induce neuroregeneration in SCI dogs, and compared this with BDNF-MSCs, HO-1-MSCs, and GFP-MSCs injected dogs. The groups were compared in terms of improvement in canine Basso, Beattie, and Bresnahan (cBBB) score during 8 weeks of experimentation. After 8 weeks, spinal cords were harvested and subjected to western blot analysis, immunofluorescent staining, and hematoxylin and eosin (H&E) staining. The combination group showed a significant improvement in hindlimb functions, with a higher BBB score, and a robust increase in neuroregeneration, depicted by a higher expression of Tuj-1, NF-M, and GAP-43 due to a decreased expression of the inflammatory markers interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α), and an increased expression of interleukin-10 (IL-10) (P ≤ 0.05). H&E staining showed more reduced intraparenchymal fibrosis in the combination group than in other groups (P ≤ 0.05). It was thus suggested that the cotransplantation of HO-1 and BDNF-MSCs is more effective in promoting the healing of SCI. HO-1-MSCs reduce inflammation, which favors BDNF-induced neuroregeneration in SCI of dogs.
Collapse
Affiliation(s)
- Imdad Ullah Khan
- 1 Department of Veterinary Surgery, College of Veterinary Medicine, Seoul National University, Gwanak-gu, Seoul, Korea
| | - Yongseok Yoon
- 1 Department of Veterinary Surgery, College of Veterinary Medicine, Seoul National University, Gwanak-gu, Seoul, Korea
| | - Ahyoung Kim
- 1 Department of Veterinary Surgery, College of Veterinary Medicine, Seoul National University, Gwanak-gu, Seoul, Korea
| | - Kwang Rae Jo
- 1 Department of Veterinary Surgery, College of Veterinary Medicine, Seoul National University, Gwanak-gu, Seoul, Korea
| | - Kyeung Uk Choi
- 1 Department of Veterinary Surgery, College of Veterinary Medicine, Seoul National University, Gwanak-gu, Seoul, Korea
| | - Taeseong Jung
- 1 Department of Veterinary Surgery, College of Veterinary Medicine, Seoul National University, Gwanak-gu, Seoul, Korea
| | - Namyul Kim
- 1 Department of Veterinary Surgery, College of Veterinary Medicine, Seoul National University, Gwanak-gu, Seoul, Korea
| | - YeonSung Son
- 1 Department of Veterinary Surgery, College of Veterinary Medicine, Seoul National University, Gwanak-gu, Seoul, Korea
| | - Wan Hee Kim
- 1 Department of Veterinary Surgery, College of Veterinary Medicine, Seoul National University, Gwanak-gu, Seoul, Korea
| | - Oh-Kyeong Kweon
- 1 Department of Veterinary Surgery, College of Veterinary Medicine, Seoul National University, Gwanak-gu, Seoul, Korea
| |
Collapse
|
30
|
Haggerty AE, Maldonado-Lasunción I, Oudega M. Biomaterials for revascularization and immunomodulation after spinal cord injury. ACTA ACUST UNITED AC 2018; 13:044105. [PMID: 29359704 DOI: 10.1088/1748-605x/aaa9d8] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Spinal cord injury (SCI) causes immediate damage to the nervous tissue accompanied by loss of motor and sensory function. The limited self-repair competence of injured nervous tissue underscores the need for reparative interventions to recover function after SCI. The vasculature of the spinal cord plays a crucial role in SCI and repair. Ruptured and sheared blood vessels in the injury epicenter and blood vessels with a breached blood-spinal cord barrier (BSCB) in the surrounding tissue cause bleeding and inflammation, which contribute to the overall tissue damage. The insufficient formation of new functional vasculature in and near the injury impedes endogenous tissue repair and limits the prospect of repair approaches. Limiting the loss of blood vessels, stabilizing the BSCB, and promoting the formation of new blood vessels are therapeutic targets for spinal cord repair. Inflammation is an integral part of injury-mediated vascular damage, which has deleterious and reparative consequences. Inflammation and the formation of new blood vessels are intricately interwoven. Biomaterials can be effectively used for promoting and guiding blood vessel formation or modulating the inflammatory response after SCI, thereby governing the extent of damage and the success of reparative interventions. This review deals with the vasculature after SCI, the reciprocal interactions between inflammation and blood vessel formation, and the potential of biomaterials to support revascularization and immunomodulation in damaged spinal cord nervous tissue.
Collapse
Affiliation(s)
- Agnes E Haggerty
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | | | | |
Collapse
|
31
|
Li M, Mei X, Lv S, Zhang Z, Xu J, Sun D, Xu J, He X, Chi G, Li Y. Rat vibrissa dermal papilla cells promote healing of spinal cord injury following transplantation. Exp Ther Med 2018; 15:3929-3939. [PMID: 29581745 PMCID: PMC5863572 DOI: 10.3892/etm.2018.5916] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Accepted: 01/18/2018] [Indexed: 12/24/2022] Open
Abstract
Bone marrow mesenchymal stem cell (BMSC) transplantation is effective for repairing spinal cord injuries (SCIs); however, there are limitations of clinical BMSC applications. Previously, we reported that dermal papilla cells (DPCs) secrete brain-derived neurotrophic factor and glial cell line-derived neurotrophic factor more actively than BMSCs. To analyze the therapeutic function of DPCs in SCI, primary DPCs and BMSCs were cultured from the same green fluorescence protein-transgenic rat. The cells were suspended in rat-tail collagen I and transplanted separately into completely transected spinal cord lesion sites. Grafted-cell survival was examined with a small animal in vivo imaging detection system, and lesion sites were examined histochemically. In vivo imaging revealed enhanced lesion filling and survival with DPC grafts compared with BMSC grafts on days 14 and 21 post-transplantation. Hematoxylin and eosin staining demonstrated that lesion area sizes in the two groups were not markedly different. In the DPC transplant group, more axons formed within the lesion sites. CD31-positive vessel-like structures were more abundant in lesion sites near the grafted cells in the DPC group. The results of the present study suggest that DPCs may be a valuable alternative source of stem cells for autologous cell therapy for the treatment of SCI.
Collapse
Affiliation(s)
- Meiying Li
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin 130021, P.R. China.,Department of Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xianglin Mei
- Department of Pathology, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China.,National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, Jilin 130024, P.R. China
| | - Shuang Lv
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Zechuan Zhang
- Clinical Medical College, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jinying Xu
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Dongjie Sun
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jiayi Xu
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xia He
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Guangfan Chi
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yulin Li
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
32
|
Liu S, Schackel T, Weidner N, Puttagunta R. Biomaterial-Supported Cell Transplantation Treatments for Spinal Cord Injury: Challenges and Perspectives. Front Cell Neurosci 2018; 11:430. [PMID: 29375316 PMCID: PMC5768640 DOI: 10.3389/fncel.2017.00430] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 12/20/2017] [Indexed: 12/17/2022] Open
Abstract
Spinal cord injury (SCI), resulting in para- and tetraplegia caused by the partial or complete disruption of descending motor and ascending sensory neurons, represents a complex neurological condition that remains incurable. Following SCI, numerous obstacles comprising of the loss of neural tissue (neurons, astrocytes, and oligodendrocytes), formation of a cavity, inflammation, loss of neuronal circuitry and function must be overcome. Given the multifaceted primary and secondary injury events that occur with SCI treatment options are likely to require combinatorial therapies. While several methods have been explored, only the intersection of two, cell transplantation and biomaterial implantation, will be addressed in detail here. Owing to the constant advance of cell culture technologies, cell-based transplantation has come to the forefront of SCI treatment in order to replace/protect damaged tissue and provide physical as well as trophic support for axonal regrowth. Biomaterial scaffolds provide cells with a protected environment from the surrounding lesion, in addition to bridging extensive damage and providing physical and directional support for axonal regrowth. Moreover, in this combinatorial approach cell transplantation improves scaffold integration and therefore regenerative growth potential. Here, we review the advances in combinatorial therapies of Schwann cells (SCs), astrocytes, olfactory ensheathing cells (OECs), mesenchymal stem cells, as well as neural stem and progenitor cells (NSPCs) with various biomaterial scaffolds.
Collapse
Affiliation(s)
- Shengwen Liu
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Thomas Schackel
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Norbert Weidner
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Radhika Puttagunta
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
33
|
Enzmann V, Lecaudé S, Kruschinski A, Vater A. CXCL12/SDF-1-Dependent Retinal Migration of Endogenous Bone Marrow-Derived Stem Cells Improves Visual Function after Pharmacologically Induced Retinal Degeneration. Stem Cell Rev Rep 2017; 13:278-286. [PMID: 27924617 DOI: 10.1007/s12015-016-9706-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Mobilized bone marrow-derived stem cells (BMSC) have been discussed as an alternative strategy for endogenous repair. Thereby, different approaches for BMSC mobilization have been pursued. Herein, the role of a newly discovered oligonucleotide for retinal homing and regeneration capability of BMSCs was investigated in the sodium iodate (NaIO3) model of retinal degeneration. Mobilization was achieved in GFP-chimera with NOX-A12, a CXC-motif chemokine ligand 12 (CXCL12)/stromal cell-derived factor 1 (SDF-1)-neutralizing L-aptamer. BMSC homing was directed by intravitreal SDF-1 injection. Visual acuity was measured using the optokinetic reflex. Paraffin cross sections were stained with hematoxylin and eosin for retinal thickness measurements. Immunohistochemistry was performed to investigate the expression of cell-specific markers after mobilization. A single dose of NOX-A12 induced significant mobilization of GFP+ cells which were found in all layers within the degenerating retina. An additional intravitreal injection of SDF-1 increased migration towards the site of injury. Thereby, the number of BMSCs (Sca-1+) found in the damaged retina increased whereas a decrease of activated microglia (Iba-1+) was found. The mobilization led to significantly increased visual acuity. However, no significant changes in retinal thickness or differentiation towards retinal cell types were detected. Systemic mobilization by a single dose of NOX-A12 showed increased homing of BMSCs into the degenerated retina, which was associated with improved visual function when injection of SDF-1 was additionally performed. The redistribution of the cells to the site of injury combined with their observed beneficial effects support the endogenous therapeutic strategy for retinal repair.
Collapse
Affiliation(s)
- Volker Enzmann
- Department of Ophthalmology, University Hospital, University of Bern, Freiburgstrasse 14, 3010, Bern, Switzerland. .,Department of Clinical Research, University of Bern, Bern, Switzerland.
| | - Stéphanie Lecaudé
- Department of Clinical Research, University of Bern, Bern, Switzerland
| | | | | |
Collapse
|
34
|
Liu S, Sandner B, Schackel T, Nicholson L, Chtarto A, Tenenbaum L, Puttagunta R, Müller R, Weidner N, Blesch A. Regulated viral BDNF delivery in combination with Schwann cells promotes axonal regeneration through capillary alginate hydrogels after spinal cord injury. Acta Biomater 2017; 60:167-180. [PMID: 28735026 DOI: 10.1016/j.actbio.2017.07.024] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 07/05/2017] [Accepted: 07/18/2017] [Indexed: 12/24/2022]
Abstract
Grafting of cell-seeded alginate capillary hydrogels into a spinal cord lesion site provides an axonal bridge while physically directing regenerating axonal growth in a linear pattern. However, without an additional growth stimulus, bridging axons fail to extend into the distal host spinal cord. Here we examined whether a combinatory strategy would support regeneration of descending axons across a cervical (C5) lateral hemisection lesion in the rat spinal cord. Following spinal cord transections, Schwann cell (SC)-seeded alginate hydrogels were grafted to the lesion site and AAV5 expressing brain-derived neurotrophic factor (BDNF) under control of a tetracycline-regulated promoter was injected caudally. In addition, we examined whether SC injection into the caudal spinal parenchyma would further enhance regeneration of descending axons to re-enter the host spinal cord. Our data show that both serotonergic and descending axons traced by biotinylated dextran amine (BDA) extend throughout the scaffolds. The number of regenerating axons is significantly increased when caudal BDNF expression is activated and transient BDNF delivery is able to sustain axons after gene expression is switched off. Descending axons are confined to the caudal graft/host interface even with continuous BDNF expression for 8weeks. Only with a caudal injection of SCs, a pathway facilitating axonal regeneration through the host/graft interface is generated allowing axons to successfully re-enter the caudal spinal cord. STATEMENT OF SIGNIFICANCE Recovery from spinal cord injury is poor due to the limited regeneration observed in the adult mammalian central nervous system. Biomaterials, cell transplantation and growth factors that can guide axons across a lesion site, provide a cellular substrate, stimulate axon growth and have shown some promise in increasing the growth distance of regenerating axons. In the present study, we combined an alginate biomaterial with linear channels with transplantation of Schwann cells within and beyond the lesion site and injection of a regulatable vector for the transient expression of brain-derived neurotrophic factor (BDNF). Our data show that only with the full combination axons extend across the lesion site and that expression of BDNF beyond 4weeks does not further increase the number of regenerating axons.
Collapse
Affiliation(s)
- Shengwen Liu
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany; Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Beatrice Sandner
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Thomas Schackel
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - LaShae Nicholson
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Abdelwahed Chtarto
- Experimental Neurosurgery Laboratory and I.R.I.B.H.M., Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Liliane Tenenbaum
- Department of Clinical Neurosciences, University Hospital of Lausanne, Lausanne, Switzerland
| | - Radhika Puttagunta
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Rainer Müller
- Department of Physical and Theoretical Chemistry, University of Regensburg, Regensburg, Germany
| | - Norbert Weidner
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Armin Blesch
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany; Stark Neurosciences Research Institute, Indiana University School of Medicine, Dept. of Neurological Surgery and Goodman Campbell Brain and Spine, Indianapolis, USA.
| |
Collapse
|
35
|
The SDF-1/CXCR4 axis promotes recovery after spinal cord injury by mediating bone marrow-derived from mesenchymal stem cells. Oncotarget 2017; 8:11629-11640. [PMID: 28099928 PMCID: PMC5355292 DOI: 10.18632/oncotarget.14619] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 12/16/2016] [Indexed: 12/27/2022] Open
Abstract
This study aims to explore the role of the SDF-1/CXCR4 axis in mediating BMSCs and SCI recovery. BMSCs were collected and SCI rat models were established. Wistar rats were assigned into the blank control, sham, SCI, SCI + BMSCs, SCI + BMSCs + SDF-1, SCI + BMSCs + AMD3100 (an inhibitor of SDF-1/CXCR4 axis) and SCI + BMSCs + SDF-1 + AMD3100 groups. Hind limb motor function was measured 7, 14, 21 and 28 days after operation. qRT-PCR, western blotting and ELISA was performed to determine the expressions of SDF-1, CXCR4, NGF, BDNF, GFAP and GAP-43, TNF-α, IL-1β, L-6 and IFN-γ. Hind limb motor function scores 7 days after the operation were reduced in the SCI rats of the blank control and sham groups. Hind limb function was found to be better in the SCI + BMSCs and SCI + BMSCs + SDF-1 groups than in the SCI, SCI + BMSCs + AMD3100 and SCI + BMSCs + SDF-1 + AMD3100 groups 14, 21 and 28 days after operation. Furthermore, the SCI group had lower SDF-1, CXCR4, NGF, BDNF and GAP-43 expressions but higher GFAP, TNF-α, IL-1β, IL-6 and IFN-γ than the blank control and sham groups 28 days after operation. While, the SCI + BMSCs, SCI + BMSCs + SDF-1 and SCI + BMSCs + SDF-1 + AMD3100 groups displayed opposite trends to the SCI and SCI + BMSCs + AMD3100 groups. In conclusion, SDF-1/CXCR4 axis promotes recovery after SCI by mediating BMSCs.
Collapse
|
36
|
Cell transplantation therapy for spinal cord injury. Nat Neurosci 2017; 20:637-647. [DOI: 10.1038/nn.4541] [Citation(s) in RCA: 607] [Impact Index Per Article: 75.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 02/22/2017] [Indexed: 02/07/2023]
|
37
|
Hodgetts SI, Harvey AR. Neurotrophic Factors Used to Treat Spinal Cord Injury. VITAMINS AND HORMONES 2016; 104:405-457. [PMID: 28215303 DOI: 10.1016/bs.vh.2016.11.007] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The application of neurotrophic factors as a therapy to improve morphological and behavioral outcomes after experimental spinal cord injury (SCI) has been the focus of many studies. These studies vary markedly in the type of neurotrophic factor that is delivered, the mode of administration, and the location, timing, and duration of the treatment. Generally, the majority of studies have had significant success if neurotrophic factors are applied in or close to the lesion site during the acute or the subacute phase after SCI. Comparatively fewer studies have administered neurotrophic factors in order to directly target the somata of injured neurons. The mode of delivery varies between acute injection of recombinant proteins, subacute or chronic delivery using a variety of strategies including osmotic minipumps, cell-mediated delivery, delivery using polymer release vehicles or supporting bridges of some sort, or the use of gene therapy to modify neurons, glial cells, or precursor/stem cells. In this brief review, we summarize the state of play of many of the therapies using these factors, most of which have been undertaken in rodent models of SCI.
Collapse
Affiliation(s)
- S I Hodgetts
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Perth, WA, Australia; Western Australian Neuroscience Research Institute, Perth, WA, Australia.
| | - A R Harvey
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Perth, WA, Australia; Western Australian Neuroscience Research Institute, Perth, WA, Australia
| |
Collapse
|
38
|
Lee SH, Kim Y, Rhew D, Kim A, Jo KR, Yoon Y, Choi KU, Jung T, Kim WH, Kweon OK. Impact of local injection of brain-derived neurotrophic factor-expressing mesenchymal stromal cells (MSCs) combined with intravenous MSC delivery in a canine model of chronic spinal cord injury. Cytotherapy 2016; 19:S1465-3249(16)30540-0. [PMID: 28029610 DOI: 10.1016/j.jcyt.2016.09.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 09/18/2016] [Accepted: 09/30/2016] [Indexed: 01/09/2023]
Abstract
BACKGROUND AIMS The microenvironment of the chronically injured spinal cord does not allow for axonal regeneration due to glial scarring. To ameliorate this, several therapeutic strategies have been used. We investigated whether combined transplantation of chondroitinase ABC (chABC) and mesenchymal stromal cells (MSCs) genetically modified to secrete brain-derived neurotrophic factor (BDNF) with intravenous (IV) administration of MSCs can promote recovery of hindlimb function after chronic spinal cord injury (SCI). METHODS Canine BDNF-expressing MSCs were generated using a lentivirus packaging protocol. Twelve beagle dogs with experimentally induced chronic SCI were divided into chABC/MSC-green fluorescent protein (GFP), chABC/MSC-BDNF and chABC/MSC-BDNF/IV groups. The MSCs (1 × 107 cells) and chABC were transplanted 3 weeks after SCI in all groups, and IV injection of MSC-GFP (1 × 107 cells) was performed 1 and 2 weeks after MSC transplantation in the chABC/MSC-BDNF/IV group. Spinal cords were harvested 8 weeks after transplantation. RESULTS The dogs in the chABC/MSC-BDNF included groups had significantly improved functional recovery 8 weeks after transplantation compared with those in the chABC/MSC-GFP group. The animals in the chABC/MSC-BDNF/IV group showed significant improvements in functional recovery at 6, 7 and 8 weeks compared with those in the chABC/MSC-BDNF group. Fibrotic changes were significantly decreased in the chABC/MSC-BDNF/IV group. We also observed significant decreases in the expression levels of tumor necrosis factor-α, interleukin-6, COX-2, glial fibrillary acidic protein and GalC and increased expression levels of BDNF, β3-tubulin neurofilament medium, and nestin in the chABC/MSC-BDNF/IV group. CONCLUSIONS We suggest that transplantation of combined chABC and BDNF-expressing MSCs, along with IV injection of MSCs, is the optimal therapy for chronic SCI.
Collapse
Affiliation(s)
- Seung Hoon Lee
- BK21 PLUS Program for Creative Veterinary Science Research, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Yongsun Kim
- BK21 PLUS Program for Creative Veterinary Science Research, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Daeun Rhew
- BK21 PLUS Program for Creative Veterinary Science Research, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Ahyoung Kim
- BK21 PLUS Program for Creative Veterinary Science Research, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Kwang Rae Jo
- BK21 PLUS Program for Creative Veterinary Science Research, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Yongseok Yoon
- BK21 PLUS Program for Creative Veterinary Science Research, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Kyeung Uk Choi
- BK21 PLUS Program for Creative Veterinary Science Research, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Taeseong Jung
- BK21 PLUS Program for Creative Veterinary Science Research, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Wan Hee Kim
- BK21 PLUS Program for Creative Veterinary Science Research, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Oh-Kyeong Kweon
- BK21 PLUS Program for Creative Veterinary Science Research, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
39
|
Brock JH, Graham L, Staufenberg E, Collyer E, Koffler J, Tuszynski MH. Bone Marrow Stromal Cell Intraspinal Transplants Fail to Improve Motor Outcomes in a Severe Model of Spinal Cord Injury. J Neurotrauma 2016; 33:1103-14. [PMID: 26414795 PMCID: PMC4904236 DOI: 10.1089/neu.2015.4009] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Bone marrow stromal cells (BMSCs) have been reported to exert potential neuroprotective properties in models of neurotrauma, although precise mechanisms underlying their benefits are poorly understood. Despite this lack of knowledge, several clinical trials have been initiated using these cells. To determine whether local mechanisms mediate BMSC neuroprotective actions, we grafted allogeneic BMSCs to sites of severe, compressive spinal cord injury (SCI) in Sprague-Dawley rats. Cells were administered 48 h after the original injury. Additional animals received allogeneic MSCs that were genetically modified to secrete brain-derived neurotrophic factor (BDNF) to further determine whether a locally administered neurotrophic factor provides or extends neuroprotection. When assessed 2 months post-injury in a clinically relevant model of severe SCI, BMSC grafts with or without BDNF secretion failed to improve motor outcomes. Thus, allogeneic grafts of BMSCs do not appear to act through local mechanisms, and future clinical trials that acutely deliver BMSCs to actual sites of injury within days are unlikely to be beneficial. Additional studies should address whether systemic administration of BMSCs alter outcomes from neurotrauma.
Collapse
Affiliation(s)
- John H. Brock
- Department of Neurosciences, University of California, San Diego, La Jolla, California
- Veterans Administration San Diego Healthcare System, La Jolla, California
| | - Lori Graham
- Department of Neurosciences, University of California, San Diego, La Jolla, California
| | - Eileen Staufenberg
- Department of Neurosciences, University of California, San Diego, La Jolla, California
| | - Eileen Collyer
- Department of Neurosciences, University of California, San Diego, La Jolla, California
| | - Jacob Koffler
- Department of Neurosciences, University of California, San Diego, La Jolla, California
| | - Mark H. Tuszynski
- Department of Neurosciences, University of California, San Diego, La Jolla, California
- Veterans Administration San Diego Healthcare System, La Jolla, California
| |
Collapse
|