1
|
Abbaszadeh ME, Esmaeili M, Bilabari M, Golchin A. Brain-derived neurotrophic factor (BDNF) as biomarker in stem cell-based therapies of preclinical spinal cord injury models: A systematic review. Tissue Cell 2025; 95:102875. [PMID: 40147167 DOI: 10.1016/j.tice.2025.102875] [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: 12/05/2024] [Revised: 03/12/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025]
Abstract
Stem cell-based therapies offer promising treatment for spinal cord injury (SCI) by reducing inflammation, restoring plasticity, and supporting neuroprotection and nerve regeneration. Brain-derived neurotrophic factor (BDNF) is crucial in SCI pathophysiology. This study reviews the impact of stem cells on BDNF expression in preclinical SCI models. A thorough search was performed in PubMed, Scopus, and Web of Science until June 2023, identifying studies on the effects of stem cells on BDNF in SCI. Two researchers reviewed and extracted data from relevant studies. This review is registered in the Prospective Register of Systematic Reviews (PROSPERO) with the registration number [CRD42023441466]. Out of 923 records, 51 studies met the inclusion criteria, involving rats (46 studies) and mice (5 studies). The contusion or compression model was used in 40 studies, and the transection model in 11. The most common stem cell types were bone marrow mesenchymal stem cells (BM-MSCs), neural stem cells (NSCs), and adipose-derived stem cells (ADSCs). BM-MSCs increased BDNF expression in 16 studies, NSCs in 9 studies, and ADSCs in only one study. This review highlights that BM-MSCs and NSCs are effective in enhancing BDNF expression in preclinical SCI models, while other stem cell types may not significantly affect BDNF levels. These findings suggest variability in the effectiveness of different stem cell therapies in modulating BDNF production for SCI treatment.
Collapse
Affiliation(s)
| | - Mahdi Esmaeili
- Department of Anatomy, Faculty of Medicine, Tabriz University of Medical Sciences, Iran
| | - Maryam Bilabari
- Department of Anatomy, Faculty of Medicine, Tabriz University of Medical Sciences, Iran
| | - Ali Golchin
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran; Department of Applied Cell Sciences, Medical School, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
2
|
Kim JW, Kim J, Lee SM, Rim YA, Sung YC, Nam Y, Kim HJ, Kim H, Jung SI, Lim J, Ju JH. Combination of induced pluripotent stem cell-derived motor neuron progenitor cells with irradiated brain-derived neurotrophic factor over-expressing engineered mesenchymal stem cells enhanced restoration of axonal regeneration in a chronic spinal cord injury rat model. Stem Cell Res Ther 2024; 15:173. [PMID: 38886817 PMCID: PMC11184802 DOI: 10.1186/s13287-024-03770-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 05/26/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Spinal cord injury (SCI) is a disease that causes permanent impairment of motor, sensory, and autonomic nervous system functions. Stem cell transplantation for neuron regeneration is a promising strategic treatment for SCI. However, selecting stem cell sources and cell transplantation based on experimental evidence is required. Therefore, this study aimed to investigate the efficacy of combination cell transplantation using the brain-derived neurotrophic factor (BDNF) over-expressing engineered mesenchymal stem cell (BDNF-eMSC) and induced pluripotent stem cell-derived motor neuron progenitor cell (iMNP) in a chronic SCI rat model. METHOD A contusive chronic SCI was induced in Sprague-Dawley rats. At 6 weeks post-injury, BDNF-eMSC and iMNP were transplanted into the lesion site via the intralesional route. At 12 weeks post-injury, differentiation and growth factors were evaluated through immunofluorescence staining and western blot analysis. Motor neuron differentiation and neurite outgrowth were evaluated by co-culturing BDNF-eMSC and iMNP in vitro in 2-dimensional and 3-dimensional. RESULTS Combination cell transplantation in the chronic SCI model improved behavioral recovery more than single-cell transplantation. Additionally, combination cell transplantation enhanced mature motor neuron differentiation and axonal regeneration at the injured spinal cord. Both BDNF-eMSC and iMNP played a critical role in neurite outgrowth and motor neuron maturation via BDNF expression. CONCLUSIONS Our results suggest that the combined transplantation of BDNF- eMSC and iMNP in chronic SCI results in a significant clinical recovery. The transplanted iMNP cells predominantly differentiated into mature motor neurons. Additionally, BDNF-eMSC exerts a paracrine effect on neuron regeneration through BDNF expression in the injured spinal cord.
Collapse
Affiliation(s)
- Jang-Woon Kim
- CiSTEM laboratory, Catholic iPSC Research Center (CiRC), College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea
- Department of Biomedicine & Health Science, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Juryun Kim
- YiPSCELL, Inc., Seoul, Republic of Korea
| | | | - Yeri Alice Rim
- CiSTEM laboratory, Catholic iPSC Research Center (CiRC), College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea
- Department of Biomedicine & Health Science, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | | | - Yoojun Nam
- YiPSCELL, Inc., Seoul, Republic of Korea
| | | | - Hyewon Kim
- YiPSCELL, Inc., Seoul, Republic of Korea
| | - Se In Jung
- CiSTEM laboratory, Catholic iPSC Research Center (CiRC), College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea
- Department of Biomedicine & Health Science, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jooyoung Lim
- CiSTEM laboratory, Catholic iPSC Research Center (CiRC), College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea
- Department of Biomedicine & Health Science, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ji Hyeon Ju
- CiSTEM laboratory, Catholic iPSC Research Center (CiRC), College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea.
- Department of Biomedicine & Health Science, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 137-701, Republic of Korea.
- YiPSCELL, Inc., Seoul, Republic of Korea.
| |
Collapse
|
3
|
Kim JW, Kim J, Mo H, Han H, Rim YA, Ju JH. Stepwise combined cell transplantation using mesenchymal stem cells and induced pluripotent stem cell-derived motor neuron progenitor cells in spinal cord injury. Stem Cell Res Ther 2024; 15:114. [PMID: 38650015 PMCID: PMC11036722 DOI: 10.1186/s13287-024-03714-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 04/02/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Spinal cord injury (SCI) is an intractable neurological disease in which functions cannot be permanently restored due to nerve damage. Stem cell therapy is a promising strategy for neuroregeneration after SCI. However, experimental evidence of its therapeutic effect in SCI is lacking. This study aimed to investigate the efficacy of transplanted cells using stepwise combined cell therapy with human mesenchymal stem cells (hMSC) and induced pluripotent stem cell (iPSC)-derived motor neuron progenitor cells (iMNP) in a rat model of SCI. METHODS A contusive SCI model was developed in Sprague-Dawley rats using multicenter animal spinal cord injury study (MASCIS) impactor. Three protocols were designed and conducted as follows: (Subtopic 1) chronic SCI + iMNP, (Subtopic 2) acute SCI + multiple hMSC injections, and (Main topic) chronic SCI + stepwise combined cell therapy using multiple preemptive hMSC and iMNP. Neurite outgrowth was induced by coculturing hMSC and iPSC-derived motor neuron (iMN) on both two-dimensional (2D) and three-dimensional (3D) spheroid platforms during mature iMN differentiation in vitro. RESULTS Stepwise combined cell therapy promoted mature motor neuron differentiation and axonal regeneration at the lesional site. In addition, stepwise combined cell therapy improved behavioral recovery and was more effective than single cell therapy alone. In vitro results showed that hMSC and iMN act synergistically and play a critical role in the induction of neurite outgrowth during iMN differentiation and maturation. CONCLUSIONS Our findings show that stepwise combined cell therapy can induce alterations in the microenvironment for effective cell therapy in SCI. The in vitro results suggest that co-culturing hMSC and iMN can synergistically promote induction of MN neurite outgrowth.
Collapse
Affiliation(s)
- Jang-Woon Kim
- CiSTEM laboratory, Catholic iPSC Research Center (CiRC), College of Medicine, The Catholic University of Korea, 06591, Seoul, Republic of Korea
- Department of Biomedicine & Health Science, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 06591, Seoul, Republic of Korea
| | | | - Hyunkyung Mo
- CiSTEM laboratory, Catholic iPSC Research Center (CiRC), College of Medicine, The Catholic University of Korea, 06591, Seoul, Republic of Korea
- Department of Biomedicine & Health Science, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 06591, Seoul, Republic of Korea
| | - Heeju Han
- CiSTEM laboratory, Catholic iPSC Research Center (CiRC), College of Medicine, The Catholic University of Korea, 06591, Seoul, Republic of Korea
- Department of Biomedicine & Health Science, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 06591, Seoul, Republic of Korea
| | - Yeri Alice Rim
- CiSTEM laboratory, Catholic iPSC Research Center (CiRC), College of Medicine, The Catholic University of Korea, 06591, Seoul, Republic of Korea.
- Department of Biomedicine & Health Science, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 06591, Seoul, Republic of Korea.
| | - Ji Hyeon Ju
- CiSTEM laboratory, Catholic iPSC Research Center (CiRC), College of Medicine, The Catholic University of Korea, 06591, Seoul, Republic of Korea.
- Department of Biomedicine & Health Science, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 06591, Seoul, Republic of Korea.
- YiPSCELL, Inc, Seoul, South Korea.
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, Institute of Medical Science, College of Medicine, The Catholic University of Korea, 06591, Seoul, Republic of Korea.
| |
Collapse
|
4
|
Wang Y, Ding Y, Guo C. Mesenchymal Stem Cells for the Treatment of Spinal Cord Injury in Rat Models: A Systematic Review and Network Meta-Analysis. Cell Transplant 2024; 33:9636897241262992. [PMID: 38910431 PMCID: PMC11265244 DOI: 10.1177/09636897241262992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 05/23/2024] [Accepted: 06/01/2024] [Indexed: 06/25/2024] Open
Abstract
Transplantation of mesenchymal stem cells (MSCs) is one of the hopeful treatments for spinal cord injury (SCI). Most current studies are in animals, and less in humans, and the optimal transplantation strategy for MSCs is still controversial. In this article, we explore the optimal transplantation strategy of MSCs through a network meta-analysis of the effects of MSCs on SCI in animal models. PubMed, Web of Science, Cochrane Library, Embase, China National Knowledge Infrastructure (CNKI), Wanfang Database, China Science and Technology Journal Database (VIP), and Chinese Biomedical Literature Service System (SinoMed) databases were searched by computer for randomized controlled studies on MSCs for SCI. Two investigators independently completed the literature screening and data extraction based on the inclusion and exclusion criteria. RevMan 5.4 software was used to assess the quality of the included literature. Stata 16.0 software was used for standard meta-analysis and network meta-analysis. Standardized mean difference (SMD) was used for continuous variables to combine the statistics and calculate 95% confidence interval (95% CI). P < 0.05 was considered a statistically significant difference. Cochrane's Q test and the I2 value were used to indicate the magnitude of heterogeneity. A random-effects model was used if I2 > 50% and P < 0.10 indicated significant heterogeneity between studies, and conversely, a fixed-effects model was used. Evidence network diagrams were drawn based on direct comparisons between various interventions. The surface under the cumulative ranking curve area (SUCRA) was used to predict the ranking of the treatment effects of each intervention. A total of 32 animal studies were included in this article for analysis. The results of the standard meta-analysis showed that MSCs improved motor ability after SCI. The network meta-analysis showed that the best treatment effect was achieved for adipose tissue-derived mesenchymal stromal cells (ADMSCs) in terms of cell source and intrathecal (IT) in terms of transplantation modality. For transplantation timing, the best treatment effect was achieved when transplantation was performed in the subacute phase. The available literature suggests that IT transplantation using ADMSCs in the subacute phase may be the best transplantation strategy to improve functional impairment after SCI. Future high-quality studies are still needed to further validate the results of this study to ensure the reliability of the results.
Collapse
Affiliation(s)
- Yueying Wang
- College of Rehabilitation Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yi Ding
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chenchen Guo
- Department of Rehabilitation Medicine, Neck, Shoulder, Lumbago and Leg Pain Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
5
|
Li DY, Li YM, Lv DY, Deng T, Zeng X, You L, Pang QY, Li Y, Zhu BM. Enhanced interaction between genome-edited mesenchymal stem cells and platelets improves wound healing in mice. J Tissue Eng 2024; 15:20417314241268917. [PMID: 39329066 PMCID: PMC11425747 DOI: 10.1177/20417314241268917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 06/26/2024] [Indexed: 09/28/2024] Open
Abstract
Impaired wound healing poses a significant burden on the healthcare system and patients. Stem cell therapy has demonstrated promising potential in the treatment of wounds. However, its clinical application is hindered by the low efficiency of cell homing. In this study, we successfully integrated P-selectin glycoprotein ligand-1 (PSGL-1) into the genome of human adipose-derived mesenchymal stem cells (ADSCs) using a Cas9-AAV6-based genome editing tool platform. Our findings revealed that PSGL-1 knock-in enhanced the binding of ADSCs to platelets and their adhesion to the injured site. Moreover, the intravenous infusion of PSGL-1 -engineered ADSCs (KI-ADSCs) significantly improved the homing efficiency and residence rate at the site of skin lesions in mice. Mechanistically, PSGL-1 knock-in promotes the release of some therapeutic cytokines by activating the canonical WNT/β-catenin signaling pathway and accelerates the healing of wounds by promoting angiogenesis, re-epithelialization, and granulation tissue formation at the wound site. This study provides a novel strategy to simultaneously address the problem of poor migration and adhesion of mesenchymal stem cells (MSCs).
Collapse
Affiliation(s)
- De-Yong Li
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yu-Meng Li
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Dan-Yi Lv
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Tian Deng
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xin Zeng
- Key Laboratory of Transplant Engineering and Immunology, National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Lu You
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qiu-Yu Pang
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yi Li
- Core Facilities of West China Hospital, Sichuan University, Chengdu, China
| | - Bing-Mei Zhu
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
6
|
Zeng CW. Advancing Spinal Cord Injury Treatment through Stem Cell Therapy: A Comprehensive Review of Cell Types, Challenges, and Emerging Technologies in Regenerative Medicine. Int J Mol Sci 2023; 24:14349. [PMID: 37762654 PMCID: PMC10532158 DOI: 10.3390/ijms241814349] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/17/2023] [Accepted: 09/19/2023] [Indexed: 09/29/2023] Open
Abstract
Spinal cord injuries (SCIs) can lead to significant neurological deficits and lifelong disability, with far-reaching physical, psychological, and economic consequences for affected individuals and their families. Current treatments for SCIs are limited in their ability to restore function, and there is a pressing need for innovative therapeutic approaches. Stem cell therapy has emerged as a promising strategy to promote the regeneration and repair of damaged neural tissue following SCIs. This review article comprehensively discusses the potential of different stem cell types, such as embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), mesenchymal stem cells (MSCs), and neural stem/progenitor cells (NSPCs), in SCI treatment. We provide an in-depth analysis of the unique advantages and challenges associated with each stem cell type, as well as the latest advancements in the field. Furthermore, we address the critical challenges faced in stem cell therapy for SCIs, including safety concerns, ethical considerations, standardization of protocols, optimization of transplantation parameters, and the development of effective outcome measures. We also discuss the integration of novel technologies such as gene editing, biomaterials, and tissue engineering to enhance the therapeutic potential of stem cells. The article concludes by emphasizing the importance of collaborative efforts among various stakeholders in the scientific community, including researchers, clinicians, bioengineers, industry partners, and patients, to overcome these challenges and realize the full potential of stem cell therapy for SCI patients. By fostering such collaborations and advancing our understanding of stem cell biology and regenerative medicine, we can pave the way for the development of groundbreaking therapies that improve the lives of those affected by SCIs.
Collapse
Affiliation(s)
- Chih-Wei Zeng
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
7
|
Yea JH, Kim Y, Jo CH. Comparison of mesenchymal stem cells from bone marrow, umbilical cord blood, and umbilical cord tissue in regeneration of a full-thickness tendon defect in vitro and in vivo. Biochem Biophys Rep 2023; 34:101486. [PMID: 37234487 PMCID: PMC10206173 DOI: 10.1016/j.bbrep.2023.101486] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/15/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Although mesenchymal stem cells (MSCs) can be obtained from various tissues such as bone marrow (BM), umbilical cord blood (UCB) and umbilical cord tissue (UC), the comparative efficacy of each MSC in tendon regeneration is unknown. Therefore, we investigated the efficacy of MSCs isolated from three different sources on tendon regeneration after injury. We evaluated the potential of BM-, UCB- and UC-MSC to differentiate into tendon-like cells in tensioned three-dimensional construct (T-3D) using gene and histological analysis. In animal experiments, full-thickness tendon defect (FTD) was created in supraspinatus of rats, and injected with Saline and BM-, UCB- and UC-MSC. After two and four weeks, histological evaluations were performed. After inducing tenogenic differentiation, the gene expression of scleraxis, mohawk, type I collagen and tenascin-C was upregulated by 3.12-, 5.92-, 6.01- and 1.61-fold respectively and formation of tendon-like matrix was increased 4.22-fold in UC-MSC compared to BM-MSC in T-3D. In animal experiments, the total degeneration score was lower in the UC-MSC group than in BM-MSC group at both weeks. In heterotopic matrix formation, glycosaminoglycan-rich area was reduced in the UC-MSC group, whereas area was larger in the BM-MSC group than in Saline group at four weeks. In conclusion, UC-MSC is superior to other MSCs in differentiating into tendon-like lineage cells and forming a well-organized tendon-like matrix under T-3D conditions. UC-MSC enhances regeneration of FTD in terms of histological properties compared to BM- and UCB-MSC.
Collapse
Affiliation(s)
- Ji-Hye Yea
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Yeasol Kim
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, South Korea
- Department of Orthopedic Surgery, SMG-SNU Boramae Medical Center, Seoul National University College of Medicine, Seoul, South Korea
| | - Chris H. Jo
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, South Korea
- Department of Orthopedic Surgery, SMG-SNU Boramae Medical Center, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
8
|
Jevans B, James ND, Burnside E, McCann CJ, Thapar N, Bradbury EJ, Burns AJ. Combined treatment with enteric neural stem cells and chondroitinase ABC reduces spinal cord lesion pathology. Stem Cell Res Ther 2021; 12:10. [PMID: 33407795 PMCID: PMC7789480 DOI: 10.1186/s13287-020-02031-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 11/16/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Spinal cord injury (SCI) presents a significant challenge for the field of neurotherapeutics. Stem cells have shown promise in replenishing the cells lost to the injury process, but the release of axon growth-inhibitory molecules such as chondroitin sulfate proteoglycans (CSPGs) by activated cells within the injury site hinders the integration of transplanted cells. We hypothesised that simultaneous application of enteric neural stem cells (ENSCs) isolated from the gastrointestinal tract, with a lentivirus (LV) containing the enzyme chondroitinase ABC (ChABC), would enhance the regenerative potential of ENSCs after transplantation into the injured spinal cord. METHODS ENSCs were harvested from the GI tract of p7 rats, expanded in vitro and characterised. Adult rats bearing a contusion injury were randomly assigned to one of four groups: no treatment, LV-ChABC injection only, ENSC transplantation only or ENSC transplantation+LV-ChABC injection. After 16 weeks, rats were sacrificed and the harvested spinal cords examined for evidence of repair. RESULTS ENSC cultures contained a variety of neuronal subtypes suitable for replenishing cells lost through SCI. Following injury, transplanted ENSC-derived cells survived and ChABC successfully degraded CSPGs. We observed significant reductions in the injured tissue and cavity area, with the greatest improvements seen in the combined treatment group. ENSC-derived cells extended projections across the injury site into both the rostral and caudal host spinal cord, and ENSC transplantation significantly increased the number of cells extending axons across the injury site. Furthermore, the combined treatment resulted in a modest, but significant functional improvement by week 16, and we found no evidence of the spread of transplanted cells to ectopic locations or formation of tumours. CONCLUSIONS Regenerative effects of a combined treatment with ENSCs and ChABC surpassed either treatment alone, highlighting the importance of further research into combinatorial therapies for SCI. Our work provides evidence that stem cells taken from the adult gastrointestinal tract, an easily accessible source for autologous transplantation, could be strongly considered for the repair of central nervous system disorders.
Collapse
Affiliation(s)
- Benjamin Jevans
- Stem Cells and Regenerative Medicine, UCL Great Ormond Street Institute of Child Health, London, UK
- Present Address: German Centre for Neurodegenerative diseases (DZNE), Bonn, Germany
| | - Nicholas D James
- Regeneration Group, The Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology & Neuroscience, King's College London, Guy's Campus, London, UK
| | - Emily Burnside
- Regeneration Group, The Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology & Neuroscience, King's College London, Guy's Campus, London, UK
| | - Conor J McCann
- Stem Cells and Regenerative Medicine, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Nikhil Thapar
- Stem Cells and Regenerative Medicine, UCL Great Ormond Street Institute of Child Health, London, UK
- Neurogastroenterology and Motility Unit, Department of Gastroenterology, Great Ormond Street Hospital, London, UK
- Present Address: Department of Paediatric Gastroenterology, Hepatology and Liver Transplant, Queensland Children's Hospital, Brisbane, Australia
| | - Elizabeth J Bradbury
- Regeneration Group, The Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology & Neuroscience, King's College London, Guy's Campus, London, UK
| | - Alan J Burns
- Stem Cells and Regenerative Medicine, UCL Great Ormond Street Institute of Child Health, London, UK.
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands.
- Present Address: Gastrointestinal Drug Discovery Unit, Takeda Pharmaceuticals International, Cambridge, USA.
| |
Collapse
|
9
|
Yea JH, Kim I, Sym G, Park JK, Lee AY, Cho BC, Bae TS, Kim BJ, Jo CH. Regeneration of a full-thickness defect in rotator cuff tendon with umbilical cord-derived mesenchymal stem cells in a rat model. PLoS One 2020; 15:e0235239. [PMID: 33166292 PMCID: PMC7652329 DOI: 10.1371/journal.pone.0235239] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 10/27/2020] [Indexed: 01/08/2023] Open
Abstract
Although rotator cuff disease is a common cause of shoulder pain, there is still no treatment method that could halt or reveres its development and progression. The purpose of this study was to investigate the efficacy of umbilical cord-derived mesenchymal stem cells (UC MSCs) on the regeneration of a full-thickness rotator cuff defect (FTD) in a rat model. We injected either UC MSCs or saline to the FTD and investigated macroscopic, histological and biomechanical results and cell trafficking. Treatment with UC MSCs improved macroscopic appearance in terms of tendon thickness at two weeks, and inflammation, defect size, swelling/redness and connection surrounding tissue and slidability at four weeks compared to the saline group. Histologically, UC MSCs induced the tendon matrix formation recovering collagen organization, nuclear aspect ratio and orientation angle of fibroblast as well as suppressing cartilage-related glycosaminoglycan compared to saline group at four weeks. The UC MSCs group also improved ultimate failure load by 25.0% and 19.0% and ultimate stress by 27.3% and 26.8% at two and four weeks compared to saline group. UC MSCs labeled with PKH26 exhibited 5.3% survival at four weeks compared to three hours after injection. This study demonstrated that UC MSCs regenerated the FTD with tendon tissue similar properties to the normal tendon in terms of macroscopic, histological and biomechanical characteristics in a rat model.
Collapse
Affiliation(s)
- Ji-Hye Yea
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
- Department of Orthopedic Surgery, SMG-SNU Boramae Medical Center, Seoul National University College of Medicine, Seoul, Korea
| | - InJa Kim
- Department of Orthopedic Surgery, SMG-SNU Boramae Medical Center, Seoul National University College of Medicine, Seoul, Korea
| | - Gayoung Sym
- Department of Orthopedic Surgery, SMG-SNU Boramae Medical Center, Seoul National University College of Medicine, Seoul, Korea
| | - Jin-Kyung Park
- Department of Orthopedic Surgery, SMG-SNU Boramae Medical Center, Seoul National University College of Medicine, Seoul, Korea
| | - Ah-Young Lee
- Department of Orthopedic Surgery, SMG-SNU Boramae Medical Center, Seoul National University College of Medicine, Seoul, Korea
| | - Byeong Chan Cho
- Department of Biomedical Engineering, Collage of Science and Engineering, Jungwon University, Goesan-gun, Chungcheongbuk-do, Korea
| | - Tae Soo Bae
- Department of Biomedical Engineering, Collage of Science and Engineering, Jungwon University, Goesan-gun, Chungcheongbuk-do, Korea
| | - Byoung Jae Kim
- Department of Obstetrics & Gynecology, SMG-SNU Boramae Medical Center, Seoul National University College of Medicine, Seoul, Korea
| | - Chris Hyunchul Jo
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
- Department of Orthopedic Surgery, SMG-SNU Boramae Medical Center, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
10
|
Vawda R, Badner A, Hong J, Mikhail M, Dragas R, Xhima K, Jose A, Fehlings MG. Harnessing the Secretome of Mesenchymal Stromal Cells for Traumatic Spinal Cord Injury: Multicell Comparison and Assessment of In Vivo Efficacy. Stem Cells Dev 2020; 29:1429-1443. [PMID: 32962528 PMCID: PMC7703247 DOI: 10.1089/scd.2020.0079] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Cell therapy offers significant promise for traumatic spinal cord injury (SCI), which despite many medical advances, has limited treatment strategies. Able to address the multifactorial and dynamic pathophysiology of SCI, cells present various advantages over standard pharmacological approaches. However, the use of live cells is also severely hampered by logistical and practical considerations. These include specialized equipment and expertise, standardization of cell stocks, sustained cell viability post-thawing, and cryopreservation-induced delayed-onset cell death. For this reason, we suggest a novel and clinically translatable alternative to live-cell systemic infusion, which retains the efficacy of the latter while overcoming many of its limitations. This strategy involves the administration of concentrated cell secretome and exploits the trophic mechanism by which stromal cells function. In this study, we compare the efficacy of intravenously delivered concentrated conditioned media (CM) from human umbilical cord matrix cells (HUCMCs), bone marrow mesenchymal stromal cells, as well as newborn and adult fibroblasts in a rat model of moderately severe cervical clip compression/contusion injury (C7--T1, 35 g). This is further paired with a thorough profile of the CM cytokines, chemokines, and angiogenic factors. The HUCMC-derived CM was most effective at limiting acute (48 h post-SCI) vascular pathology, specifically lesion volume, and functional vascularity. Principle component analysis (PCA), hierarchical clustering, and interaction analysis of proteins highly expressed in the HUCMC secretome suggest involvement of the MAPK/ERK, JAK/STAT, and immune cell migratory pathways. This "secretotherapeutic" strategy represents a novel and minimally invasive method to target multiple organ systems and several pathologies shortly after traumatic SCI.
Collapse
Affiliation(s)
- Reaz Vawda
- Department of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Canada
| | - Anna Badner
- Department of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Canada.,Institute of Medical Science, School of Medicine, University of Toronto, Toronto, Canada
| | - James Hong
- Department of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Canada.,Institute of Medical Science, School of Medicine, University of Toronto, Toronto, Canada
| | - Mirriam Mikhail
- Department of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Canada
| | - Rachel Dragas
- Department of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Canada.,Institute of Medical Science, School of Medicine, University of Toronto, Toronto, Canada
| | - Kristiana Xhima
- Department of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Canada
| | - Alejandro Jose
- Department of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Canada
| | - Michael G Fehlings
- Department of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Canada.,Institute of Medical Science, School of Medicine, University of Toronto, Toronto, Canada.,Spinal Program, Toronto Western Hospital, University Health Network, Toronto, Canada
| |
Collapse
|
11
|
Regeneration of the rotator cuff tendon-to-bone interface using umbilical cord-derived mesenchymal stem cells and gradient extracellular matrix scaffolds from adipose tissue in a rat model. Acta Biomater 2020; 114:104-116. [PMID: 32682057 DOI: 10.1016/j.actbio.2020.07.020] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 07/07/2020] [Accepted: 07/09/2020] [Indexed: 12/17/2022]
Abstract
Regeneration of the gradient structure of the tendon-to-bone interface (TBI) is a crucial goal after rotator cuff repair. The purpose of this study was to investigate the efficacy of a biomimetic hydroxyapatite-gradient scaffold (HA-G scaffold) isolated from adipose tissue (AD) with umbilical cord derived mesenchymal stem cells (UC MSCs) on the regeneration of the structure of the TBI by analyzing the histological and biomechanical changes in a rat repair model. As a result, the HA-G scaffold had progressively increased numbers of hydroxyapatite (HA) particles from the tendon to the bone phase. After seeding UC MSCs to the scaffold, specific matrices, such as collagen, glycoaminoglycan, and calcium, were synthesized with respect to the HA density. In a rat repair model, compared to the repair group, the UC MSCs seeded HA-G scaffold group had improved collagen organization and cartilage formation by 52% at 8 weeks and 262.96% at 4 weeks respectively. Moreover, ultimate failure load also increased by 30.71% at 4 weeks in the UC MSCs seeded HA-G scaffold group compared to the repair group. Especially, the improved values were comparable to values in normal tissue. This study demonstrated that HA-G scaffold isolated from AD induced UC MSCs to form tendon, cartilage and bone matrices similar to the TBI structure according to the HA density. Furthermore, UC MSC-seeded HA-G scaffold regenerated the TBI of the rotator cuff in a rat repair model in terms of histological and biomechanical properties similar to the normal TBI. Statement of Significance We found specific extracellular matrix (ECM) formation in the biomimetic-hydroxyapatite-gradient-scaffold (HA-G-scaffold) in vitro as well as improved histological and biomechanical results of repaired rotator cuff after the scaffold implantation in a rat model. This study has four strengths; An ECM scaffold derived from human adipose tissue; only one-layer used for a gradient scaffold not a multilayer used to mimic the unique structure of the gradient tendon-to-bone-interface (TBI) of the rotator cuff; UC-MSCs as a new cell source for TBI regeneration; and the UC-MSCs synthesized specific matrices with respect to the HA density without any other stimuli. This study suggested that the UC-MSC seeded HA-G-scaffold could be used as a promising strategy for the regeneration of rotator cuff tears.
Collapse
|
12
|
Yea JH, Park JK, Kim IJ, Sym G, Bae TS, Jo CH. Regeneration of a full-thickness defect of rotator cuff tendon with freshly thawed umbilical cord-derived mesenchymal stem cells in a rat model. Stem Cell Res Ther 2020; 11:387. [PMID: 32894193 PMCID: PMC7487485 DOI: 10.1186/s13287-020-01906-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 08/05/2020] [Accepted: 08/26/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND It is difficult to immediately use mesenchymal stem cells (MSCs) for the patient with rotator cuff disease because isolation and culture time are required. Thus, the MSCs would be prepared in advanced in cryopreserved condition for an "off-the-shelf" usage in clinic. This study investigated the efficacy of freshly thawed MSCs on the regeneration of a full-thickness tendon defect (FTD) of rotator cuff tendon in a rat model. METHODS We evaluated morphology, viability, and proliferation of cultured umbilical cord-derived MSCs (C-UC MSCs) and freshly thawed umbilical cord-derived MSCs (T-UC MSCs) at passage 10 in vitro. In animal experiments, we created a FTD in the supraspinatus of rats and injected the injured tendon with saline, cryopreserved agent (CPA; control), C-UC MSCs, and T-UC MSCs, respectively. Two and 4 weeks later, macroscopic, histological, biomechanical, and cell trafficking were evaluated. T test and ANOVA were used with SPSS. Differences with p < .05 were considered statistically significant. RESULTS T-UC MSCs had fibroblast-like morphology and showed greater than 97% viability and stable proliferation comparable to the C-UC MSCs at passage 10. In animal experiments, compared with the control group, the macroscopic appearance of the T-UC MSCs was more recovered at 2 and 4 weeks such as inflammation, defect size, neighboring tendon, swelling/redness, the connecting surrounding tissue and slidability. Histologically, the nuclear aspect ratio, orientation angle of fibroblasts, collagen organization, and fiber coherence were improved by 33.33%, 42.75%, 1.86-fold, and 1.99-fold at 4 weeks, and GAG-rich area decreased by 88.13% and 94.70% at 2 and 4 weeks respectively. Further, the T-UC MSCs showed enhanced ultimate failure load by 1.55- and 1.25-fold compared with the control group at both 2 and 4 weeks. All the improved values of T-UC MSCs were comparable to those of C-UC MSCs. Moreover, T-UC MSCs remained 8.77% at 4 weeks after injury, and there was no significant difference between C-UC MSCs and T-UC MSCs. CONCLUSIONS The morphology, viability, and proliferation of T-UC MSCs were comparable to those of C-UC MSCs. Treatment with T-UC MSCs could induce tendon regeneration of FTD at the macroscopic, histological, and biomechanical levels comparable to treatment with C-UC MSCs.
Collapse
Affiliation(s)
- Ji-Hye Yea
- Department of Translational Medicine, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
- Department of Orthopedic Surgery, SMG-SNU Boramae Medical Center, Seoul National University College of Medicine, 20 Boramae-ro 5-gil, Dongjak-gu, Seoul, 07061, Korea
| | - Jin-Kyung Park
- Department of Orthopedic Surgery, SMG-SNU Boramae Medical Center, Seoul National University College of Medicine, 20 Boramae-ro 5-gil, Dongjak-gu, Seoul, 07061, Korea
| | - In Ja Kim
- Department of Orthopedic Surgery, SMG-SNU Boramae Medical Center, Seoul National University College of Medicine, 20 Boramae-ro 5-gil, Dongjak-gu, Seoul, 07061, Korea
| | - Gayoung Sym
- Department of Orthopedic Surgery, SMG-SNU Boramae Medical Center, Seoul National University College of Medicine, 20 Boramae-ro 5-gil, Dongjak-gu, Seoul, 07061, Korea
| | - Tae-Soo Bae
- Department of Biomedical Engineering, Collage of Science and Engineering, Jungwon University, 85, Munmu-ro, Goesan-eup, Goesan-gun, Chungcheongbuk-do, 367-805, Korea
| | - Chris Hyunchul Jo
- Department of Translational Medicine, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Korea.
- Department of Orthopedic Surgery, SMG-SNU Boramae Medical Center, Seoul National University College of Medicine, 20 Boramae-ro 5-gil, Dongjak-gu, Seoul, 07061, Korea.
| |
Collapse
|
13
|
Hakim R, Covacu R, Zachariadis V, Frostell A, Sankavaram SR, Brundin L, Svensson M. Mesenchymal stem cells transplanted into spinal cord injury adopt immune cell-like characteristics. Stem Cell Res Ther 2019; 10:115. [PMID: 30944028 PMCID: PMC6448247 DOI: 10.1186/s13287-019-1218-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 02/27/2019] [Accepted: 03/20/2019] [Indexed: 02/06/2023] Open
Abstract
Background Mesenchymal stem cells (MSCs) and their cellular response to various stimuli have been characterized in great detail in culture conditions. In contrast, the cellular response of MSCs in an in vivo setting is still uncharted territory. In this study, we investigated the cellular response of MSCs following transplantation into spinal cord injury (SCI). Methods Mouse bone marrow-derived MSCs were transplanted 24 h following severe contusion SCI in mice. As controls, MSCs transplanted to the uninjured spinal cord and non-transplanted MSCs were used. At 7 days post transplantation, the MSCs were isolated from the SCI, and their global transcriptional changes, survival, differentiation, proliferation, apoptosis, and phenotypes were investigated using RNA sequencing, immunohistochemistry, and flow cytometry. Results MSCs transplanted into SCI downregulated genes related to cell-cycle regulation/progression, DNA metabolic/biosynthetic process, and DNA repair and upregulated genes related to immune system response, cytokine production/response, response to stress/stimuli, signal transduction and signaling pathways, apoptosis, and phagocytosis/endocytosis. MSCs maintained their surface expression of Sca1 and CD29 but upregulated expression of CD45 following transplantation. Transplanted MSCs maintained their surface expression of MHC-I but upregulated surface expression of MHC-II. Transplanted MSCs survived and proliferated to a low extent, did not express Caspase-3, and did not differentiate into neurons or astrocytes. Conclusion MSCs transplanted into SCI upregulate expression of CD45 and MHC-II and expression of genes related to cytokine production, phagocytosis/endocytosis, and immune cells/response and thereby adopt immune cell-like characteristics within the recipient. Electronic supplementary material The online version of this article (10.1186/s13287-019-1218-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ramil Hakim
- Department of Neurology, Karolinska University Hospital, 17176, Stockholm, Sweden.,Department of Clinical Neuroscience, Karolinska Institutet, 17176, Stockholm, Sweden.,BioClinicum, Karolinska University Hospital, Solnavägen 30, Solna, 171 64, Stockholm, Sweden
| | - Ruxandra Covacu
- Department of Neurology, Karolinska University Hospital, 17176, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institutet, 17176, Stockholm, Sweden.,Department of Clinical Neuroscience, Karolinska Institutet, 17176, Stockholm, Sweden
| | - Vasilios Zachariadis
- Department of Oncology and Pathology, Karolinska Institutet, 17176, Stockholm, Sweden
| | - Arvid Frostell
- Department of Clinical Neuroscience, Karolinska Institutet, 17176, Stockholm, Sweden.,BioClinicum, Karolinska University Hospital, Solnavägen 30, Solna, 171 64, Stockholm, Sweden.,Department of Neurosurgery, Karolinska University Hospital, 17176, Stockholm, Sweden
| | - Sreenivasa Raghavan Sankavaram
- Center for Molecular Medicine, Karolinska Institutet, 17176, Stockholm, Sweden.,Department of Clinical Neuroscience, Karolinska Institutet, 17176, Stockholm, Sweden
| | - Lou Brundin
- Department of Neurology, Karolinska University Hospital, 17176, Stockholm, Sweden. .,Department of Clinical Neuroscience, Karolinska Institutet, 17176, Stockholm, Sweden. .,BioClinicum, Karolinska University Hospital, Solnavägen 30, Solna, 171 64, Stockholm, Sweden.
| | - Mikael Svensson
- Department of Clinical Neuroscience, Karolinska Institutet, 17176, Stockholm, Sweden.,BioClinicum, Karolinska University Hospital, Solnavägen 30, Solna, 171 64, Stockholm, Sweden.,Department of Neurosurgery, Karolinska University Hospital, 17176, Stockholm, Sweden
| |
Collapse
|
14
|
Effects of Different Doses of Mesenchymal Stem Cells on Functional Recovery After Compressive Spinal-Cord Injury in Mice. Neuroscience 2019; 400:17-32. [DOI: 10.1016/j.neuroscience.2018.12.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 12/03/2018] [Accepted: 12/04/2018] [Indexed: 02/07/2023]
|
15
|
Jevans B, McCann CJ, Thapar N, Burns AJ. Transplanted enteric neural stem cells integrate within the developing chick spinal cord: implications for spinal cord repair. J Anat 2018; 233:592-606. [PMID: 30191559 DOI: 10.1111/joa.12880] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/10/2018] [Indexed: 12/27/2022] Open
Abstract
Spinal cord injury (SCI) causes paralysis, multisystem impairment and reduced life expectancy, as yet with no cure. Stem cell therapy can potentially replace lost neurons, promote axonal regeneration and limit scar formation, but an optimal stem cell source has yet to be found. Enteric neural stem cells (ENSC) isolated from the enteric nervous system (ENS) of the gastrointestinal (GI) tract are an attractive source. Here, we used the chick embryo to assess the potential of ENSC to integrate within the developing spinal cord. In vitro, isolated ENSC formed extensive cell connections when co-cultured with spinal cord (SC)-derived cells. Further, qRT-PCR analysis revealed the presence of TuJ1+ neurons, S100+ glia and Sox10+ stem cells within ENSC neurospheres, as well as expression of key neuronal subtype genes, at levels comparable to SC tissue. Following ENSC transplantation to an ablated region of chick embryo SC, donor neurons were found up to 12 days later. These neurons formed bridging connections within the SC injury zone, aligned along the anterior/posterior axis, and were immunopositive for TuJ1. These data provide early proof of principle support for the use of ENSCs for SCI, and encourage further research into their potential for repair.
Collapse
Affiliation(s)
- Benjamin Jevans
- Stem Cells and Regenerative Medicine, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Conor J McCann
- Stem Cells and Regenerative Medicine, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Nikhil Thapar
- Stem Cells and Regenerative Medicine, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Alan J Burns
- Stem Cells and Regenerative Medicine, UCL Great Ormond Street Institute of Child Health, London, UK.,Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands.,Gastrointestinal Drug Discovery Unit, Takeda Pharmaceuticals International, Cambridge, MA, USA
| |
Collapse
|
16
|
Naderi-Meshkin H, Ahmadiankia N. Cancer metastasis versus stem cell homing: Role of platelets. J Cell Physiol 2018; 233:9167-9178. [PMID: 30105746 DOI: 10.1002/jcp.26937] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 06/12/2018] [Indexed: 12/12/2022]
Abstract
One of the major obstacles in achieving a successful stem cell therapy is insufficient homing of transplanted cells. To overcome this obstacle, understanding the underlying mechanisms of stem cell homing is of obvious importance. Central to this review is the concept that cancer metastasis can be viewed as a role model to build up a comprehensive concept of stem cell homing. In this novel perspective, the prosurvival choices of the cancerous cells in the bloodstream, their arrest, extravasation, and proliferation at the secondary site can be exploited in favor of targeted stem cell homing. To date, tumor cells have been found to employ a wide variety of strategies to promote metastasis. One of these strategies is through their ability to activate platelets and subsequently activated platelets serve cancer cell survival and metastasis. Accordingly, in the first part of this review the roles of platelets in cancer metastasis as well as stem cell homing are discussed. Next, we provide some lessons learned from cancer metastasis in favor of developing strategies for improvement of stem cell homing with emphasis on the role of platelets. Based on direct or indirect evidence from metastasis, strategies such as manipulation of stem cells to enhance interaction with platelets, preconditioning-pretreatment of stem cells with platelets in vitro, and coinjection of both stem cells and platelets are proposed to improve stem cell homing.
Collapse
Affiliation(s)
- Hojjat Naderi-Meshkin
- Stem Cells and Regenerative Medicine Research Group, Iranian Academic Center for Education, Culture Research (ACECR), Khorasan Razavi Branch, Mashhad, Iran
| | - Naghmeh Ahmadiankia
- School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran.,Cancer Prevention Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
| |
Collapse
|
17
|
Mesenchymal Stem Cell-Based Therapy Improves Lower Limb Movement After Spinal Cord Ischemia in Rats. Ann Thorac Surg 2018; 105:1523-1530. [DOI: 10.1016/j.athoracsur.2017.12.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 10/28/2017] [Accepted: 12/12/2017] [Indexed: 11/19/2022]
|
18
|
Mesenchymal Stem Cells Form 3D Clusters Following Intraventricular Transplantation. J Mol Neurosci 2018; 65:60-73. [PMID: 29705933 DOI: 10.1007/s12031-018-1070-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 04/19/2018] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells (MSCs) are regarded as an immune privileged cell type with numerous regeneration-promoting effects. The in vivo behavior of MSC and underlying mechanisms leading to their regenerative effects are largely unknown. The aims of this study were to comparatively investigate the in vivo behavior of canine (cMSC), human (hMSC), and murine MSC (mMSC) following intra-cerebroventricular transplantation. At 7 days post transplantation (dpt), clusters of cMSC, hMSC, and mMSC were detected within the ventricular system. At 49 dpt, cMSC-transplanted mice showed clusters mostly consisting of extracellular matrix lacking transplanted MSC. Similarly, hMSC-transplanted mice lacked MSC clusters at 49 dpt. Xenogeneic MSC transplantation was associated with a local T lymphocyte-dominated immune reaction at both time points. Interestingly, no associated inflammation was observed following syngeneic mMSC transplantation. In conclusion, transplanted MSC formed intraventricular cell clusters and exhibited a short life span in vivo. Xenogeneically in contrast to syngeneically transplanted MSC triggered a T cell-mediated graft rejection indicating that MSCs are not as immune privileged as previously assumed. However, MSC may mediate their effects by a "hit and run" mechanism and future studies will show whether syngeneically or xenogeneically transplanted MSCs exert better therapeutic effects in animals with CNS disease.
Collapse
|
19
|
Hosseini SM, Sani M, Haider KH, Dorvash M, Ziaee SM, Karimi A, Namavar MR. Concomitant use of mesenchymal stem cells and neural stem cells for treatment of spinal cord injury: A combo cell therapy approach. Neurosci Lett 2018; 668:138-146. [PMID: 29317311 DOI: 10.1016/j.neulet.2018.01.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 01/03/2018] [Accepted: 01/04/2018] [Indexed: 12/16/2022]
Abstract
with neural stem cells (NSCs) provides a hope to recover the neural damage and compensate for the lost neural structures for restoration of interrupted neural communications above and below the site of injury. However, cell-based therapy approach suffers from many biological barriers and technical caveats which severely hamper the prognosis. The biochemically-rich microenvironment at the site of spinal cord injury (SCI), the continuing neuro-degenerative process and infiltrating immune cells offer a serious barrier to the donor cells. We hypothesized that mesenchymal stem cells (MSCs) concomitantly delivered with NSCs would significantly enhance the effectiveness of cell-based therapy for SCI. In a rodent model of SCI (n = 15 animals/group), MSCs labeled with PKH67 (green fluorescence dye) were delivered on day1 after SCI whereas the same animals were treated with NSCs during the subacute phase on day3 (group-5). In comparison with untreated control (group-1), sham group (without cell treatment; group-2), MSCs alone (group-3) and NSCs alone treated animals (group-4), the combined cell treated animals (group-5) showed significantly higher homing of cells at the site of injury during in vivo imaging. Caspase-3 activity was lower in group-5 (P < 0.05 vs all groups) with concomitant reduction in the pro-inflammatory cytokines IL-1β and IL-6 (P < 0.05 vs all groups). All cell therapy groups showed significant improvement in neurological function as compared to group-2, however, it was highest in group-5 (P < 0.05 vs all groups). In conclusion, combined treatment with (NSCs + MSCs) enhances NSCs survival and functional recovery in SCI and is superior to the treatment with either of NSCs or MSCs alone.
Collapse
Affiliation(s)
- Seyed Mojtaba Hosseini
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran; Cell & Molecular Medicine Student Research Group, Medical Faculty, Shiraz University of Medical Sciences, Shiraz, Iran; Stem Cell Laboratory, Department of Anatomy, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mahsa Sani
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran; Cell & Molecular Medicine Student Research Group, Medical Faculty, Shiraz University of Medical Sciences, Shiraz, Iran; Stem Cell Laboratory, Department of Anatomy, Shiraz University of Medical Sciences, Shiraz, Iran.
| | | | - Mohammadreza Dorvash
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Seyyed Mohyeddin Ziaee
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran; Cell & Molecular Medicine Student Research Group, Medical Faculty, Shiraz University of Medical Sciences, Shiraz, Iran; Stem Cell Laboratory, Department of Anatomy, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Aliasghar Karimi
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran.
| | - Mohmmad Reza Namavar
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Anatomy, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
20
|
Induction of Endogenous Neural Stem Cells By Extracorporeal Shock Waves After Spinal Cord Injury. Spine (Phila Pa 1976) 2018; 43:E200-E207. [PMID: 28658046 DOI: 10.1097/brs.0000000000002302] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
STUDY DESIGN Animal experimental study OBJECTIVES.: The purpose of this study is to investigate the effects of extracorporeal shock waves (ESWs) on endogenous neural stem cells (NSCs) proliferation after spinal cord injury (SCI). SUMMARY OF BACKGROUND DATA Exogenous stem cell transplantation for SCI still has many limitations to be addressed such as ideal cell sources, timing of transplantation, and fate of the transplanted cells. Moreover, the efficacy is another issue owing to a peculiar pathologic condition in the chronic phase of SCI. METHODS Contusive SCI was made using 24 Sprague-Dawley rats, and ESWs were applied at post-injury 4 weeks in rats. Proliferation and differentiation of endogenous NSCs (DCX, Sox-2) and axonal sprouting (GAP-43 and MAP-2) were observed at 6 weeks after application of ESWs. Differentiation of the activated neural stem cells was also investigated by coexpression of neuronal/glial cell markers (GFAP, Neu N, and CC-1). Immunofluorescence staining and western blotting were performed for quantitative analysis, and these results were compared with those in the control group. For clinical assessment, the BBB locomotor rating scale was performed. RESULTS More proliferation of endogenous neural stem cells was noted in the experimental groups, and these activated cells were mainly founded in the ependymal layer of the central canal and the injured posterior horn. Differentiation into neuronal and glial cells was also noted in a limited number of cells. With respect to axonal regeneration, GAP-43 and MAP-2 expressions in the experimental groups were also significantly higher than those in the control group. During 6 weeks' clinical observation following ESWs application, functional improvement of the hindlimb was observed without clinical deterioration by trials. CONCLUSION Collectively, these findings indicate that ESWs on the chronic phase of SCI induce activation of endogenous NSCs and consequent functional improvement. LEVEL OF EVIDENCE N/A.
Collapse
|
21
|
Ramalho BDS, Almeida FMD, Sales CM, de Lima S, Martinez AMB. Injection of bone marrow mesenchymal stem cells by intravenous or intraperitoneal routes is a viable alternative to spinal cord injury treatment in mice. Neural Regen Res 2018; 13:1046-1053. [PMID: 29926832 PMCID: PMC6022457 DOI: 10.4103/1673-5374.233448] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In spite of advances in surgical care and rehabilitation, the consequences of spinal cord injury (SCI) are still challenging. Several experimental therapeutic strategies have been studied in the SCI field, and recent advances have led to the development of therapies that may act on the inhibitory microenvironment. Assorted lineages of stem cells are considered a good treatment for SCI. This study investigated the effect of systemic transplantation of mesenchymal stem cells (MSCs) in a compressive SCI model. Here we present results of the intraperitoneal route, which has not been used previously for MSC administration after compressive SCI. We used adult female C57BL/6 mice that underwent laminectomy at the T9 level, followed by spinal cord compression for 1 minute with a 30-g vascular clip. The animals were divided into five groups: sham (anesthesia and laminectomy but without compression injury induction), MSC i.p. (intraperitoneal injection of 8 × 105 MSCs in 500 µL of DMEM at 7 days after SCI), MSC i.v. (intravenous injection of 8 × 105 MSCs in 500 µL of DMEM at 7 days after SCI), DMEM i.p. (intraperitoneal injection of 500 µL of DMEM at 7 days after SCI), DMEM i.v. (intravenous injection of 500 µL of DMEM at 7 days after SCI). The effects of MSCs transplantation in white matter sparing were analyzed by luxol fast blue staining. The number of preserved fibers was counted in semithin sections stained with toluidine blue and the presence of trophic factors was analyzed by immunohistochemistry. In addition, we analyzed the locomotor performance with Basso Mouse Scale and Global Mobility Test. Our results showed white matter preservation and a larger number of preserved fibers in the MSC groups than in the DMEM groups. Furthermore, the MSC groups had higher levels of trophic factors (brain-derived neurotrophic factor, nerve growth factor, neurotrophin-3 and neurotrophin-4) in the spinal cord and improved locomotor performance. Our results indicate that injection of MSCs by either intraperitoneal or intravenous routes results in beneficial outcomes and can be elected as a choice for SCI treatment.
Collapse
Affiliation(s)
- Bruna Dos Santos Ramalho
- Laboratório de Neurodegeneração e Reparo, Departamento de Patologia - Faculdade de Medicina, HUCFF, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fernanda Martins de Almeida
- Laboratório de Neurodegeneração e Reparo, Departamento de Patologia - Faculdade de Medicina, HUCFF, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Conrado Mendonça Sales
- Laboratório de Neurodegeneração e Reparo, Departamento de Patologia - Faculdade de Medicina, HUCFF, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Silmara de Lima
- Laboratório de Neurodegeneração e Reparo, Departamento de Patologia - Faculdade de Medicina, HUCFF, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Maria Blanco Martinez
- Laboratório de Neurodegeneração e Reparo, Departamento de Patologia - Faculdade de Medicina, HUCFF, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
22
|
Kim YC, Kim YH, Kim JW, Ha KY. Transplantation of Mesenchymal Stem Cells for Acute Spinal Cord Injury in Rats: Comparative Study between Intralesional Injection and Scaffold Based Transplantation. J Korean Med Sci 2016; 31:1373-82. [PMID: 27510379 PMCID: PMC4974177 DOI: 10.3346/jkms.2016.31.9.1373] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 05/13/2016] [Indexed: 01/01/2023] Open
Abstract
Experimental stem cell therapy for spinal cord injury (SCI) has been extensively investigated. The selection of effective cell transplantation route is also an important issue. Although various types of scaffold have been widely tried as a carrier of stem cells to the injured spinal cord, there was little comparative study to investigate the efficacy of transplantation comparing with conventional transplantation route. A total of 48 Sprague-Dawley rats were subjected to standardized SCI, followed by transplantation of allogeneic mesenchymal stem cells (MSCs), either via intralesional injection (IL group), or via the poly (lactic-co-glycolic acid) (PLGA) scaffold (IP group) or chitosan scaffold (IC group). Engraftment and differentiation of the transplanted cells, expression of neurotrophic factors in the injured spinal cord, and functional recovery were compared with those of the control group. The mean numbers of engrafted MSCs in the IL, IP, and IC groups were 20.6 ± 0.7, 25.6 ± 1.7 and 26.7 ± 1.8 cells/high power filed (HPF), respectively. Results showed higher success rate of MSCs engraftment in the scaffold groups compared to the IL group. Expression of neuroprotective growth factors in the SCI lesions showed no significant differences between the IL, IP, and IC groups. The mean Basso, Beattie and Bresnahan locomotor scales at 6 weeks post-transplantation in the IL, IP, IC, and control groups were 7.9 ± 1.1, 7.9 ± 2.1, 8.7 ± 2.1, and 2.9 ± 1.0, respectively. The functional improvement was most excellent in the IC group. The scaffold based MSC transplantation for acute SCI presented the better cell engraftment and neuroprotective effect compared to the intralesional injection transplantation.
Collapse
Affiliation(s)
- Yoon Chung Kim
- Department of Orthopaedic Surgery, The Catholic University of Korea, St. Vincent's Hospital, Suwon, Korea
| | - Young Hoon Kim
- Department of Orthopaedic Surgery, The Catholic University of Korea, Seoul St. Mary's Hospital, Seoul, Korea
| | - Jang Woon Kim
- Department of Orthopaedic Surgery, The Catholic University of Korea, Seoul St. Mary's Hospital, Seoul, Korea
| | - Kee Yong Ha
- Department of Orthopaedic Surgery, The Catholic University of Korea, Seoul St. Mary's Hospital, Seoul, Korea.
| |
Collapse
|
23
|
Effects of Magnetically Guided, SPIO-Labeled, and Neurotrophin-3 Gene-Modified Bone Mesenchymal Stem Cells in a Rat Model of Spinal Cord Injury. Stem Cells Int 2015; 2016:2018474. [PMID: 26649047 PMCID: PMC4663356 DOI: 10.1155/2016/2018474] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 03/30/2015] [Accepted: 03/31/2015] [Indexed: 12/12/2022] Open
Abstract
Despite advances in our understanding of spinal cord injury (SCI) mechanisms, there are still no effective treatment approaches to restore functionality. Although many studies have demonstrated that transplanting NT3 gene-transfected bone marrow-derived mesenchymal stem cells (BMSCs) is an effective approach to treat SCI, the approach is often low efficient in the delivery of engrafted BMSCs to the site of injury. In this study, we investigated the therapeutic effects of magnetic targeting of NT3 gene-transfected BMSCs via lumbar puncture in a rat model of SCI. With the aid of a magnetic targeting cells delivery system, we can not only deliver the engrafted BMSCs to the site of injury more efficiently, but also perform cells imaging in vivo using MR. In addition, we also found that this composite strategy could significantly improve functional recovery and nerve regeneration compared to transplanting NT3 gene-transfected BMSCs without magnetic targeting system. Our results suggest that this composite strategy could be promising for clinical applications.
Collapse
|
24
|
Oliver-Vila I, Coca MI, Grau-Vorster M, Pujals-Fonts N, Caminal M, Casamayor-Genescà A, Ortega I, Reales L, Pla A, Blanco M, García J, Vives J. Evaluation of a cell-banking strategy for the production of clinical grade mesenchymal stromal cells from Wharton's jelly. Cytotherapy 2015; 18:25-35. [PMID: 26549383 DOI: 10.1016/j.jcyt.2015.10.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 09/15/2015] [Accepted: 10/02/2015] [Indexed: 01/09/2023]
Abstract
BACKGROUND AIMS Umbilical cord (UC) has been proposed as a source of mesenchymal stromal cells (MSCs) for use in experimental cell-based therapies provided that its collection does not raise any risk to the donor, and, similar to bone marrow and lipoaspirates, UC-MSCs are multipotent cells with immuno-modulative properties. However, some of the challenges that make a broader use of UC-MSCs difficult include the limited availability of fresh starting tissue, time-consuming processing for successful derivation of cell lines, and the lack of information on identity, potency and genetic stability in extensively expanded UC-MSCs, which are necessary for banking relevant cell numbers for preclinical and clinical studies. METHODS Factors affecting the success of the derivation process (namely, time elapsed from birth to processing and weight of fragments), and methods for establishing a two-tiered system of Master Cell Bank and Working Cell Bank of UC-MSCs were analyzed. RESULTS Efficient derivation of UC-MSCs was achieved by using UC fragments larger than 7 g that were processed within 80 h from birth. Cells maintained their immunophenotype (being highly positive for CD105, CD90 and CD73 markers), multi-potentiality and immuno-modulative properties beyond 40 cumulative population doublings. No genetic abnormalities were found, as determined by G-banding karyotype, human telomerase reverse transcriptase activity was undetectable and no toxicity was observed in vivo after intravenous administration of UC-MSCs in athymic rats. DISCUSSION This works demonstrates the feasibility of the derivation and large-scale expansion of UC-MSCs from small and relatively old fragments of UC typically discarded from public cord blood banking programs.
Collapse
Affiliation(s)
- Irene Oliver-Vila
- Divisió de Teràpies Avançades/XCELIA, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005 Barcelona, Spain
| | - Maria Isabel Coca
- Divisió de Teràpies Avançades/XCELIA, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005 Barcelona, Spain
| | - Marta Grau-Vorster
- Divisió de Teràpies Avançades/XCELIA, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005 Barcelona, Spain
| | - Noèlia Pujals-Fonts
- Divisió de Teràpies Avançades/XCELIA, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005 Barcelona, Spain
| | - Marta Caminal
- Divisió de Teràpies Avançades/XCELIA, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005 Barcelona, Spain
| | - Alba Casamayor-Genescà
- Divisió de Teràpies Avançades/XCELIA, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005 Barcelona, Spain
| | - Isabel Ortega
- Divisió de Teràpies Avançades/XCELIA, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005 Barcelona, Spain
| | - Laura Reales
- Divisió de Teràpies Avançades/XCELIA, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005 Barcelona, Spain
| | - Arnau Pla
- Divisió de Teràpies Avançades/XCELIA, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005 Barcelona, Spain
| | - Margarita Blanco
- Divisió de Teràpies Avançades/XCELIA, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005 Barcelona, Spain
| | - Joan García
- Divisió de Teràpies Avançades/XCELIA, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005 Barcelona, Spain
| | - Joaquim Vives
- Divisió de Teràpies Avançades/XCELIA, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005 Barcelona, Spain.
| |
Collapse
|
25
|
Histological study on the role of bone marrow-derived mesenchymal stem cells on the sciatic nerve and the gastrocnemius muscle in a model of sciatic nerve crush injury in albino rats. ACTA ACUST UNITED AC 2015. [DOI: 10.1097/01.ehx.0000470653.67231.07] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
26
|
Sabapathy V, Tharion G, Kumar S. Cell Therapy Augments Functional Recovery Subsequent to Spinal Cord Injury under Experimental Conditions. Stem Cells Int 2015; 2015:132172. [PMID: 26240569 PMCID: PMC4512598 DOI: 10.1155/2015/132172] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 02/04/2015] [Accepted: 02/05/2015] [Indexed: 02/06/2023] Open
Abstract
The spinal cord injury leads to enervation of normal tissue homeostasis ultimately leading to paralysis. Until now there is no proper cure for the treatment of spinal cord injury. Recently, cell therapy in animal spinal cord injury models has shown some progress of recovery. At present, clinical trials are under progress to evaluate the efficacy of cell transplantation for the treatment of spinal cord injury. Different types of cells such as pluripotent stem cells derived neural cells, mesenchymal stromal cells, neural stem cells, glial cells are being tested in various spinal cord injury models. In this review we highlight both the advances and lacuna in the field of spinal cord injury by discussing epidemiology, pathophysiology, molecular mechanism, and various cell therapy strategies employed in preclinical and clinical injury models and finally we discuss the limitations and ethical issues involved in cell therapy approach for treating spinal cord injury.
Collapse
Affiliation(s)
- Vikram Sabapathy
- Centre for Stem Cell Research, Christian Medical College, Bagayam, Vellore, Tamil Nadu 632002, India
| | - George Tharion
- Department of Physical Medicine and Rehabilitation, Christian Medical College, Vellore, Tamil Nadu 632002, India
| | - Sanjay Kumar
- Centre for Stem Cell Research, Christian Medical College, Bagayam, Vellore, Tamil Nadu 632002, India
| |
Collapse
|
27
|
Dong BT, Tu GJ, Han YX, Chen Y. Lithium enhanced cell proliferation and differentiation of mesenchymal stem cells to neural cells in rat spinal cord. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:2473-2483. [PMID: 26045753 PMCID: PMC4440062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 02/22/2015] [Indexed: 06/04/2023]
Abstract
Lithium has been shown to inhibit apoptosis of neural progenitor cells (NPCs) and promote differentiation of NPCs. However, there was rare data to discuss the effects of lithium on neural differentiation of mesenchymal stem cells (MSCs). Here, we investigated the potential promotion of lithium to MSC proliferation and neural differentiation in vitro and after transplanted into the ventral horn of rat spinal cord in vivo. We found that lithium possesses the ability to promote proliferation of GFP-MSCs in a dose dependent manner as verified by growth curve and bromodeoxyuridine (BrdU) incorporation assays; While in neural induction medium, lithium (0.1 mM) promotes neural differentiation of GFP-MSCs as verified by immunostaining and quantitative analysis. After transplantation of GFP-MSCs into the rat spinal cord, lithium treatment enhanced cell survival and neural differentiation after transplantation as verified by immunohistochemistry. These data suggested that lithium could be a potential drug to augment the therapeutic efficiency of MSCs transplantation therapy in central nervous system (CNS) disorders.
Collapse
Affiliation(s)
- Bao-Tie Dong
- Department of Orthopaedic, First Affiliated Hospital of China Medical University Shenyang 110001, China
| | - Guan-Jun Tu
- Department of Orthopaedic, First Affiliated Hospital of China Medical University Shenyang 110001, China
| | - Ya-Xin Han
- Department of Orthopaedic, First Affiliated Hospital of China Medical University Shenyang 110001, China
| | - Yi Chen
- Department of Orthopaedic, First Affiliated Hospital of China Medical University Shenyang 110001, China
| |
Collapse
|
28
|
Abstract
Stem cell-based interventions aim to use special regenerative cells (stem cells) to facilitate neuronal function beyond the site of the injury. Many studies involving animal models of spinal cord injury (SCI) suggest that certain stem cell-based therapies may restore function after SCI. Currently, in case of spinal cord injuries, new discoveries with clinical implications have been continuously made in basic stem cell research, and stem cell-based approaches are advancing rapidly toward application in patients. There is a huge base of preclinical evidence in vitro and in animal models which suggests the safety and clinical efficacy of cellular therapies after SCI. Despite this, data from clinical studies is not very encouraging and at times confounding. Here, we have attempted to cover preclinical and clinical evidence base dealing with safety, feasibility and efficacy of cell based interventions after SCI. The limitations of preclinical data and the reasons underlying its failure to translate in a clinical setting are also discussed. Based on the evidence base, it is suggested that a multifactorial approach is required to address this situation. Need for standardized, stringently designed multi-centric clinical trials for obtaining validated proof of evidence is also highlighted.
Collapse
Affiliation(s)
- Harvinder Singh Chhabra
- Spine Service, Indian Spinal Injuries Centre, Vasant Kunj, New Delhi, India,Address for correspondence: Dr. Harvinder Singh Chhabra, Indian Spinal Injuries Centre, Sector C, Vasant Kunj, New Delhi - 110 070, India. E-mail:
| | - Kanchan Sarda
- Spine Service, Indian Spinal Injuries Centre, Vasant Kunj, New Delhi, India
| |
Collapse
|
29
|
Does extracorporeal shock wave introduce alteration of microenvironment in cell therapy for chronic spinal cord injury? Spine (Phila Pa 1976) 2014; 39:E1553-9. [PMID: 25271504 DOI: 10.1097/brs.0000000000000626] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
STUDY DESIGN Animal experimental study. OBJECTIVE To present experimental evidence for cell therapy for spinal cord injury (SCI). SUMMARY OF BACKGROUND DATA In chronic SCI, the efficacy of cell engraftment has been known to be low due to its distinct pathology. Alteration of microenvironment was tried using extracorporeal shock waves (ESW) for chronic SCI, and the efficacy of cell therapy was investigated. METHODS A chronic contusive SCI model was made in 36 Sprague-Dawley rats. The rats were allocated into (1) control group (SCI only), (2) ESW control group (SCI + ESW), (3) IV group (SCI + intravenous transplantation of mesenchymal stem cells; MSCs), and (4) ESW + IV group (SCI + MSCs IV transplantation after ESW). ESW were applied at the energy determined by our preliminary trials. Engraftment of the cells and expressions of growth factors (brain-derived neurotrophic factor, neuronal growth factor) and cytokines (SDF-1, CXCR4, VEGF) at the epicenter were assessed. The Basso, Beattie, and Bresnahan locomotor scale was used for the clinical assessment. RESULTS The mean numbers of engrafted cells were higher in the ESW+ IV than that in the IV with a statistical significance. The expression of SDF-1 was higher in the ESW groups than that in the control or IV group. CXCR4 was highly expressed in the transplanted groups. The expressions of growth factors in the treated group were higher in the treated group than those in the control group. However, various statistical significances were noted. The improvement of locomotor was higher in the transplanted groups than that in the control and ESW only group. CONCLUSION At a given energy level, ESW presented more engraftment of the transplanted MSCs without any clinical deterioration in a chronic SCI. Based on this promising result and possible explanations, ESW may cause an alteration of the microenvironment for the cell therapy in chronic SCI. LEVEL OF EVIDENCE N/A.
Collapse
|
30
|
Sarmento CAP, Rodrigues MN, Bocabello RZ, Mess AM, Miglino MA. Pilot study: bone marrow stem cells as a treatment for dogs with chronic spinal cord injury. Regen Med Res 2014; 2:9. [PMID: 25984337 PMCID: PMC4422475 DOI: 10.1186/2050-490x-2-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 10/15/2014] [Indexed: 01/08/2023] Open
Abstract
Background Chronic Spinal Cord injury is a common, severe, and medically untreatable disease. Since the functional outcomes of acute and experimental chronic spinal cord injury have been shown to improve with stem cell therapy, a case study was conducted to test if the application of stem cell also regenerates chronic SCI dysfunction. Transplantation of foetal bone marrow stem cells was applied in seven dogs with chronic spinal cord injury. Magnetic resonance images and assessments of symptoms according to the Olby scale were used to diagnose the severity of injury. Result All dogs improved locomotor and sensory function when examined 90 days after surgery, and showed increased movement of the hind limbs, and were able to stand upright, as well as to take small steps. Tail tone was observed in seven dogs, pain reflexes and defecation return were observed in five dogs. Conclusion The transplantation of bone marrow stem may be a promising, reliable and safe treatment for chronic spinal cord injury. Electronic supplementary material The online version of this article (doi:10.1186/2050-490X-2-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Marcio Nogueira Rodrigues
- School of Veterinary Medicine and Animal Sciences, University of São Paulo, São Paulo, Brazil ; Cidade Universitaria-Butanta, Av. Prof. Orlando Marques de Paiva, 87, São Paulo, 05508270 Brazil
| | | | - Andrea Maria Mess
- School of Veterinary Medicine and Animal Sciences, University of São Paulo, São Paulo, Brazil
| | - Maria Angelica Miglino
- School of Veterinary Medicine and Animal Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
31
|
Zhang C, Lv G. Repair of sciatic nerve defects using tissue engineered nerves. Neural Regen Res 2014; 8:1985-94. [PMID: 25206507 PMCID: PMC4145905 DOI: 10.3969/j.issn.1673-5374.2013.21.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Accepted: 06/17/2013] [Indexed: 01/24/2023] Open
Abstract
In this study, we constructed tissue-engineered nerves with acellular nerve allografts in Sprague-Dawley rats, which were prepared using chemical detergents-enzymatic digestion and mechanical methods, in combination with bone marrow mesenchymal stem cells of Wistar rats cultured in vitro, to repair 15 mm sciatic bone defects in Wistar rats. At postoperative 12 weeks, electrophysiological detection results showed that the conduction velocity of regenerated nerve after repair with tissue-engineered nerves was similar to that after autologous nerve grafting, and was higher than that after repair with acellular nerve allografts. Immunohistochemical staining revealed that motor endplates with acetylcholinesterase-positive nerve fibers were orderly arranged in the middle and superior parts of the gastrocnemius muscle; regenerated nerve tracts and sprouted branches were connected with motor endplates, as shown by acetylcholinesterase histochemistry combined with silver staining. The wet weight ratio of the tibialis anterior muscle at the affected contralateral hind limb was similar to the sciatic nerve after repair with autologous nerve grafts, and higher than that after repair with acellular nerve allografts. The hind limb motor function at the affected side was significantly improved, indicating that acellular nerve allografts combined with bone marrow mesenchymal stem cell bridging could promote functional recovery of rats with sciatic nerve defects.
Collapse
Affiliation(s)
- Caishun Zhang
- Department of Hand and Foot Surgery, First Affiliated Hospital of Liaoning Medical University, Jinzhou 121001, Liaoning Province, China
| | - Gang Lv
- Department of Hand and Foot Surgery, First Affiliated Hospital of Liaoning Medical University, Jinzhou 121001, Liaoning Province, China
| |
Collapse
|
32
|
Abstract
Stem cells have emerged as promising tools for the treatment of incurable neural and heart diseases and tissue damage. However, the survival of transplanted stem cells is reported to be low, reducing their therapeutic effects. The major causes of poor survival of stem cells in vivo are linked to anoikis, potential immune rejection, and oxidative damage mediating apoptosis. This review investigates novel methods and potential molecular mechanisms for stem cell preconditioning in vitro to increase their retention after transplantation in damaged tissues. Microenvironmental preconditioning (e.g., hypoxia, heat shock, and exposure to oxidative stress), aggregate formation, and hydrogel encapsulation have been revealed as promising strategies to reduce cell apoptosis in vivo while maintaining biological functions of the cells. Moreover, this review seeks to identify methods of optimizing cell dose preparation to enhance stem cell survival and therapeutic function after transplantation.
Collapse
Affiliation(s)
- Sébastien Sart
- Hydrodynamics Laboratory , CNRS UMR7646, Ecole Polytechnique, Palaiseau, France
| | - Teng Ma
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University , Tallahassee, Florida
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University , Tallahassee, Florida
| |
Collapse
|
33
|
Wang LJ, Zhang RP, Li JD. Transplantation of neurotrophin-3-expressing bone mesenchymal stem cells improves recovery in a rat model of spinal cord injury. Acta Neurochir (Wien) 2014; 156:1409-18. [PMID: 24744011 DOI: 10.1007/s00701-014-2089-6] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 03/27/2014] [Indexed: 12/11/2022]
Abstract
BACKGROUND This study aimed to investigate the therapeutic effects of transplanting neutrophin-3 (NT-3)-expressing bone marrow-derived mesenchymal stem cells (BMSCs) in a rat model of spinal cord injury (SCI). METHODS Forty-eight adult female Sprague-Dawley rats were randomly assigned to three groups: the control, BMSC, and NT-3-BMSC groups. BMSCs were infected with NT-3-DsRed or DsRed lentivirus and injected into the cerebrospinal fluid (CSF) via lumbar puncture (LP) 7 days after SCI in the NT-3-BMSC and BMSC groups, respectively. The hind-limb motor function of all rats was recorded using the Basso, Beattie, and Bresnahan (BBB) locomotor rating scale on days 1, 3, 7, 14, 21, 28, and 35 after transplantation. Haematoxylin-eosin (HE) staining, immunofluorescence labelling, and western blotting were performed at the final time point. RESULTS Expressions of NT-3, brain-derived neurotrophic factor (BDNF), and vascular endothelial growth factor (VEGF) proteins increased significantly in the NT-3-BMSC group, and hind-limb locomotor functions improved significantly in the NT-3-BMSC group compared with the other two groups. The cystic cavity area was smallest in the NT-3-BMSC group. In the NT-3-BMSC group, neurofilament 200 (NF200) and glial fibrillary acidic protein (GFAP) expression levels around the lesions were significantly increased and decreased, respectively. CONCLUSIONS Our findings demonstrate that transplantation of NT-3 gene-modified BMSCs via LP can strengthen the therapeutic benefits of BMSC transplantation. We observed that these modified cells increased locomotor function recovery, promoted nerve regeneration, and improved the injured spinal cord microenvironment, suggesting that it could be a promising treatment for SCI.
Collapse
Affiliation(s)
- Ling-Jie Wang
- Department of Medical Imaging, Shanxi Medical University, Taiyuan, People's Republic of China,
| | | | | |
Collapse
|
34
|
Zhang LX, Yin YM, Zhang ZQ, Deng LX. Grafted bone marrow stromal cells: a contributor to glial repair after spinal cord injury. Neuroscientist 2014; 21:277-89. [PMID: 24777423 DOI: 10.1177/1073858414532171] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In the CNS, astrocytes, oligodendrocytes and microglias are involved in not only development but also pathology such as spinal cord injury (SCI). Glial cells play dual roles (negative vs. positive effects) in these processes. After SCI, detrimental effects usually dominate and significantly retard functional recovery, and curbing these effects is critical for promoting neurological improvement. Bone marrow stromal cells (BMSCs) represent a new therapeutic approach for SCI by enabling improved sensory and motor functions in animal models. Although transdifferentiation to spinal neurons was poor, because of their pleiotropic nature, the protective effects of BMSCs are broad and are primarily mediated through modulation of transdifferentiation into host spinal glial components. Transplantation of BMSCs can positively alter the spinal microenvironment and enhance recovery. The objective of this review is to discuss these and other related mechanisms. Since BMSCs transplantation has been applied in other clinical fields, we hope to provide useful clues for the clinical application of BMSCs to treat the SCI in the near future.
Collapse
Affiliation(s)
- Li-Xin Zhang
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yan-Mei Yin
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Zhi-Qiang Zhang
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Ling-Xiao Deng
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, and Department of Neurological Surgery, Goodman Campbell Brain and Spine, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
35
|
Martinez AMB, Goulart CDO, Ramalho BDS, Oliveira JT, Almeida FM. Neurotrauma and mesenchymal stem cells treatment: From experimental studies to clinical trials. World J Stem Cells 2014; 6:179-94. [PMID: 24772245 PMCID: PMC3999776 DOI: 10.4252/wjsc.v6.i2.179] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 02/26/2014] [Accepted: 03/11/2014] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cell (MSC) therapy has attracted the attention of scientists and clinicians around the world. Basic and pre-clinical experimental studies have highlighted the positive effects of MSC treatment after spinal cord and peripheral nerve injury. These effects are believed to be due to their ability to differentiate into other cell lineages, modulate inflammatory and immunomodulatory responses, reduce cell apoptosis, secrete several neurotrophic factors and respond to tissue injury, among others. There are many pre-clinical studies on MSC treatment for spinal cord injury (SCI) and peripheral nerve injuries. However, the same is not true for clinical trials, particularly those concerned with nerve trauma, indicating the necessity of more well-constructed studies showing the benefits that cell therapy can provide for individuals suffering the consequences of nerve lesions. As for clinical trials for SCI treatment the results obtained so far are not as beneficial as those described in experimental studies. For these reasons basic and pre-clinical studies dealing with MSC therapy should emphasize the standardization of protocols that could be translated to the clinical set with consistent and positive outcomes. This review is based on pre-clinical studies and clinical trials available in the literature from 2010 until now. At the time of writing this article there were 43 and 36 pre-clinical and 19 and 1 clinical trials on injured spinal cord and peripheral nerves, respectively.
Collapse
Affiliation(s)
- Ana Maria Blanco Martinez
- Ana Maria Blanco Martinez, Camila de Oliveira Goulart, Bruna dos Santos Ramalho, Júlia Teixeira Oliveira, Fernanda Martins Almeida, Laboratory of Neurodegeneration and Repair, Institute of Biomedical Sciences, Health Science Center, 21941-902, Rio de Janeiro, Brazil
| | - Camila de Oliveira Goulart
- Ana Maria Blanco Martinez, Camila de Oliveira Goulart, Bruna dos Santos Ramalho, Júlia Teixeira Oliveira, Fernanda Martins Almeida, Laboratory of Neurodegeneration and Repair, Institute of Biomedical Sciences, Health Science Center, 21941-902, Rio de Janeiro, Brazil
| | - Bruna Dos Santos Ramalho
- Ana Maria Blanco Martinez, Camila de Oliveira Goulart, Bruna dos Santos Ramalho, Júlia Teixeira Oliveira, Fernanda Martins Almeida, Laboratory of Neurodegeneration and Repair, Institute of Biomedical Sciences, Health Science Center, 21941-902, Rio de Janeiro, Brazil
| | - Júlia Teixeira Oliveira
- Ana Maria Blanco Martinez, Camila de Oliveira Goulart, Bruna dos Santos Ramalho, Júlia Teixeira Oliveira, Fernanda Martins Almeida, Laboratory of Neurodegeneration and Repair, Institute of Biomedical Sciences, Health Science Center, 21941-902, Rio de Janeiro, Brazil
| | - Fernanda Martins Almeida
- Ana Maria Blanco Martinez, Camila de Oliveira Goulart, Bruna dos Santos Ramalho, Júlia Teixeira Oliveira, Fernanda Martins Almeida, Laboratory of Neurodegeneration and Repair, Institute of Biomedical Sciences, Health Science Center, 21941-902, Rio de Janeiro, Brazil
| |
Collapse
|
36
|
Dedeepiya VD, William JB, Parthiban JKBC, Chidambaram R, Balamurugan M, Kuroda S, Iwasaki M, Preethy S, Abraham SJK. The known-unknowns in spinal cord injury, with emphasis on cell-based therapies - a review with suggestive arenas for research. Expert Opin Biol Ther 2014; 14:617-34. [PMID: 24660978 DOI: 10.1517/14712598.2014.889676] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION In spite of extensive research, the progress toward a cure in spinal cord injury (SCI) is still elusive, which holds good for the cell- and stem cell-based therapies. We have critically analyzed seven known gray areas in SCI, indicating the specific arenas for research to improvise the outcome of cell-based therapies in SCI. AREAS COVERED The seven, specific known gray areas in SCI analyzed are: i) the gap between animal models and human victims; ii) uncertainty about the time, route and dosage of cells applied; iii) source of the most efficacious cells for therapy; iv) inability to address the vascular compromise during SCI; v) lack of non-invasive methodologies to track the transplanted cells; vi) need for scaffolds to retain the cells at the site of injury; and vii) physical and chemical stimuli that might be required for synapses formation yielding functional neurons. EXPERT OPINION Further research on scaffolds for retaining the transplanted cells at the lesion, chemical and physical stimuli that may help neurons become functional, a meta-analysis of timing of the cell therapy, mode of application and larger clinical studies are essential to improve the outcome.
Collapse
Affiliation(s)
- Vidyasagar Devaprasad Dedeepiya
- Nichi-In Centre for Regenerative Medicine (NCRM), The Mary-Yoshio Translational Hexagon (MYTH) , PB 1262, Chennai - 600034, Tamil Nadu , India +91 44 24732186 ; ,
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Neuroprotective effects of adipose-derived stem cells are maintained for 3 weeks against ischemic damage in the rabbit spinal cord. BIOMED RESEARCH INTERNATIONAL 2014; 2014:539051. [PMID: 24592394 PMCID: PMC3925622 DOI: 10.1155/2014/539051] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Accepted: 12/16/2013] [Indexed: 11/17/2022]
Abstract
In the previous study, we demonstrated that adipose-derived stem cells (ASCs) have neuroprotective effects against ischemic damage in the ventral horn of L5-6 levels at 3 days after ischemia/reperfusion. In the present study, we expanded our observations for 3 weeks after ischemia/reperfusion to rule out the possibility of delayed neuronal death in several days after ischemia/reperfusion. Transient spinal cord ischemia was induced by a 15 min aortic artery occlusion in the subrenal region and rabbit ASCs were administered intrathecally into recipient rabbits (2 × 105) immediately after reperfusion. Transplantation of ASCs improved the neurological motor functions of the hindlimb 3 weeks after ischemia/reperfusion. Similarly, the cresyl violet-positive neurons were significantly increased at 3 weeks after ischemia/reperfusion compared to that in the vehicle (artificial cerebrospinal fluid)-treated group. The transplantation of ASCs significantly reduced reactive microglia induced by ischemia at 3 weeks after ischemia/reperfusion. In addition, transplantation of ASCs maintained the brain-derived neurotrophic factor (BDNF) levels 3 weeks after ischemia/reperfusion. These results suggest that the neuroprotective effects of ASCs are maintained 3 weeks after ischemia/reperfusion by modulating microgliosis and BDNF levels in the spinal cord.
Collapse
|
38
|
Intravenous transplantation of mesenchymal stromal cells to enhance peripheral nerve regeneration. BIOMED RESEARCH INTERNATIONAL 2013; 2013:573169. [PMID: 24459671 PMCID: PMC3888686 DOI: 10.1155/2013/573169] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 10/07/2013] [Accepted: 10/07/2013] [Indexed: 12/16/2022]
Abstract
Peripheral nerve injury is a common and devastating complication after trauma and can cause irreversible impairment or even complete functional loss of the affected limb. While peripheral nerve repair results in some axonal regeneration and functional recovery, the clinical outcome is not optimal and research continues to optimize functional recovery after nerve repair. Cell transplantation approaches are being used experimentally to enhance regeneration. Intravenous infusion of mesenchymal stromal cells (MSCs) into spinal cord injury and stroke was shown to improve functional outcome. However, the repair potential of intravenously transplanted MSCs in peripheral nerve injury has not been addressed yet. Here we describe the impact of intravenously infused MSCs on functional outcome in a peripheral nerve injury model. Rat sciatic nerves were transected followed, by intravenous MSCs transplantation. Footprint analysis was carried out and 21 days after transplantation, the nerves were removed for histology. Labelled MSCs were found in the sciatic nerve lesion site after intravenous injection and regeneration was improved. Intravenously infused MSCs after acute peripheral nerve target the lesion site and survive within the nerve and the MSC treated group showed greater functional improvement. The results of study suggest that nerve repair with cell transplantation could lead to greater functional outcome.
Collapse
|
39
|
Kaminski EL, Falavigna A, Venturin GT, Marinowic D, Bagatini PB, Xavier LL, DaCosta JC. Two intrathecal transplants of bone marrow mononuclear cells produce motor improvement in an acute and severe model of spinal cord injury. COLUNA/COLUMNA 2013. [DOI: 10.1590/s1808-18512013000400001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
OBJECTIVE: We studied transplants of bone marrow mononuclear cells (BMMC) by lumbar puncture (LP) in a severe model of spinal cord injury (SCI) using clip compression. METHODS: BMMCs or saline solution were transplanted by LP 48 hours and 9 days post injury. Motor function was evaluated by BBB scale, histological analysis by Nissl technique and the verification of cell migration by PCR analysis. RESULTS: The BBB had significantly improved in rats treated with BMMCs by LP compared with controls (p<0.001). The histological analysis did not showed difference in the lesional area between the groups. The PCR analysis was able to found BMMCs in the injury site. CONCLUSIONS: two BMMC transplants by LP improved motor function in a severe model of SCI and BMMC was found in the injury site.
Collapse
Affiliation(s)
| | | | | | - Daniel Marinowic
- Pontifícia Universidade Católica do Rio Grande do Sul, Brasil; Pontifícia Universidade Católica do Rio Grande do Sul, Brasil
| | | | | | | |
Collapse
|
40
|
Oliveri RS, Bello S, Biering-Sørensen F. Mesenchymal stem cells improve locomotor recovery in traumatic spinal cord injury: systematic review with meta-analyses of rat models. Neurobiol Dis 2013; 62:338-53. [PMID: 24148857 DOI: 10.1016/j.nbd.2013.10.014] [Citation(s) in RCA: 112] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Revised: 09/13/2013] [Accepted: 10/10/2013] [Indexed: 12/13/2022] Open
Abstract
Traumatic spinal cord injury (SCI) is a devastating event with huge personal and societal costs. A limited number of treatments exist to ameliorate the progressive secondary damage that rapidly follows the primary mechanical impact. Mesenchymal stem or stromal cells (MSCs) have anti-inflammatory and neuroprotective effects and may thus reduce secondary damage after administration. We performed a systematic review with quantitative syntheses to assess the evidence of MSCs versus controls for locomotor recovery in rat models of traumatic SCI, and identified 83 eligible controlled studies comprising a total of 1,568 rats. Between-study heterogeneity was large. Fifty-three studies (64%) were reported as randomised, but only four reported adequate methodologies for randomisation. Forty-eight studies (58%) reported the use of a blinded outcome assessment. A random-effects meta-analysis yielded a difference in behavioural Basso-Beattie-Bresnahan (BBB) locomotor score means of 3.9 (95% confidence interval [CI] 3.2 to 4.7; P<0.001) in favour of MSCs. Trial sequential analysis confirmed the findings of the meta-analyses with the upper monitoring boundary for benefit being crossed by the cumulative Z-curve before reaching the diversity-adjusted required information size. Only time from intervention to last follow-up remained statistically significant after adjustment using multivariate random-effects meta-regression modelling. Lack of other demonstrable explanatory variables could be due to insufficient meta-analytic study power. MSCs would seem to demonstrate a substantial beneficial effect on locomotor recovery in a widely-used animal model of traumatic SCI. However, the animal results should be interpreted with caution concerning the internal and external validity of the studies in relation to the design of future clinical trials.
Collapse
Affiliation(s)
- Roberto S Oliveri
- Cell Therapy Facility, The Blood Bank, Department of Clinical Immunology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark.
| | - Segun Bello
- The Nordic Cochrane Centre, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Fin Biering-Sørensen
- Department of Spinal Cord Injuries, Copenhagen University Hospital Rigshospitalet and Glostrup Hospital, Copenhagen, Denmark
| |
Collapse
|
41
|
Cantinieaux D, Quertainmont R, Blacher S, Rossi L, Wanet T, Noël A, Brook G, Schoenen J, Franzen R. Conditioned medium from bone marrow-derived mesenchymal stem cells improves recovery after spinal cord injury in rats: an original strategy to avoid cell transplantation. PLoS One 2013; 8:e69515. [PMID: 24013448 PMCID: PMC3754952 DOI: 10.1371/journal.pone.0069515] [Citation(s) in RCA: 171] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 06/10/2013] [Indexed: 12/11/2022] Open
Abstract
Spinal cord injury triggers irreversible loss of motor and sensory functions. Numerous strategies aiming at repairing the injured spinal cord have been studied. Among them, the use of bone marrow-derived mesenchymal stem cells (BMSCs) is promising. Indeed, these cells possess interesting properties to modulate CNS environment and allow axon regeneration and functional recovery. Unfortunately, BMSC survival and differentiation within the host spinal cord remain poor, and these cells have been found to have various adverse effects when grafted in other pathological contexts. Moreover, paracrine-mediated actions have been proposed to explain the beneficial effects of BMSC transplantation after spinal cord injury. We thus decided to deliver BMSC-released factors to spinal cord injured rats and to study, in parallel, their properties in vitro. We show that, in vitro, BMSC-conditioned medium (BMSC-CM) protects neurons from apoptosis, activates macrophages and is pro-angiogenic. In vivo, BMSC-CM administered after spinal cord contusion improves motor recovery. Histological analysis confirms the pro-angiogenic action of BMSC-CM, as well as a tissue protection effect. Finally, the characterization of BMSC-CM by cytokine array and ELISA identified trophic factors as well as cytokines likely involved in the beneficial observed effects. In conclusion, our results support the paracrine-mediated mode of action of BMSCs and raise the possibility to develop a cell-free therapeutic approach.
Collapse
Affiliation(s)
- Dorothée Cantinieaux
- GIGA-Neuroscience, Axonal Regeneration and Cephalic Pain Unit, University of Liege, Liege, Belgium
| | - Renaud Quertainmont
- GIGA-Neuroscience, Axonal Regeneration and Cephalic Pain Unit, University of Liege, Liege, Belgium
| | - Silvia Blacher
- GIGA-Cancer, Laboratory of Biology of Tumour and Development, University of Liege, Liege, Belgium
| | - Loïc Rossi
- GIGA-Neuroscience, Axonal Regeneration and Cephalic Pain Unit, University of Liege, Liege, Belgium
| | - Thomas Wanet
- GIGA-Neuroscience, Axonal Regeneration and Cephalic Pain Unit, University of Liege, Liege, Belgium
| | - Agnès Noël
- GIGA-Cancer, Laboratory of Biology of Tumour and Development, University of Liege, Liege, Belgium
| | - Gary Brook
- Department of Neuropathology, University of Aachen, Aachen, Germany
| | - Jean Schoenen
- GIGA-Neuroscience, Axonal Regeneration and Cephalic Pain Unit, University of Liege, Liege, Belgium
| | - Rachelle Franzen
- GIGA-Neuroscience, Axonal Regeneration and Cephalic Pain Unit, University of Liege, Liege, Belgium
| |
Collapse
|
42
|
Cho H, Choi YK, Lee DH, Park HJ, Seo YK, Jung H, Kim SC, Kim SM, Park JK. Effects of magnetic nanoparticle-incorporated human bone marrow-derived mesenchymal stem cells exposed to pulsed electromagnetic fields on injured rat spinal cord. Biotechnol Appl Biochem 2013; 60:596-602. [DOI: 10.1002/bab.1109] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 02/13/2013] [Indexed: 11/09/2022]
Affiliation(s)
- Hyunjin Cho
- Research Institute of Biotechnology; Dongguk University; Seoul Korea
| | - Yun-Kyong Choi
- Department of Medical Biotechnology; Dongguk University; Seoul Korea
| | - Dong Heon Lee
- Advanced Functional Nanohybrid Material Lab, Department of Chemistry; Dongguk University; Seoul Korea
| | - Hee Jung Park
- Department of Medical Biotechnology; Dongguk University; Seoul Korea
| | - Young-Kwon Seo
- Research Institute of Biotechnology; Dongguk University; Seoul Korea
- Department of Medical Biotechnology; Dongguk University; Seoul Korea
| | - Hyun Jung
- Advanced Functional Nanohybrid Material Lab, Department of Chemistry; Dongguk University; Seoul Korea
- Department of Energy and Materials Engineering; Dongguk University; Seoul Korea
| | - Soo-Chan Kim
- Graduate School of Bio & Information Technology; Hankyong National University; Anseong-si Kyonggi-do Korea
| | - Sung-Min Kim
- Department of Medical Biotechnology; Dongguk University; Seoul Korea
| | - Jung-Keug Park
- Research Institute of Biotechnology; Dongguk University; Seoul Korea
- Department of Medical Biotechnology; Dongguk University; Seoul Korea
| |
Collapse
|
43
|
De Vocht N, Praet J, Reekmans K, Le Blon D, Hoornaert C, Daans J, Berneman Z, Van der Linden A, Ponsaerts P. Tackling the physiological barriers for successful mesenchymal stem cell transplantation into the central nervous system. Stem Cell Res Ther 2013; 4:101. [PMID: 23998480 PMCID: PMC3854758 DOI: 10.1186/scrt312] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Over the past decade a lot of research has been performed towards the therapeutic use of mesenchymal stem cells (MSCs) in neurodegenerative and neuroinflammatory diseases. MSCs have shown to be beneficial in different preclinical studies of central nervous system (CNS) disorders due to their immunomodulatory properties and their capacity to secrete various growth factors. Nevertheless, most of the transplanted cells die within the first hours after transplantation and induce a neuroinflammatory response. In order to increase the efficacy of MSC transplantation, it is thus imperative to completely characterise the mechanisms mediating neuroinflammation and cell death following MSC transplantation into the CNS. Consequently, different components of these cell death- and neuroinflammation-inducing pathways can be targeted in an attempt to improve the therapeutic potential of MSCs for CNS disorders.
Collapse
|
44
|
Bone marrow-derived mesenchymal stem cell transplantation for chronic spinal cord injury in rats: comparative study between intralesional and intravenous transplantation. Spine (Phila Pa 1976) 2013; 38:E1065-74. [PMID: 23629485 DOI: 10.1097/brs.0b013e31829839fa] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
STUDY DESIGN Animal experimental study. OBJECTIVE To present experimental evidence for mesenchymal cell therapy for spinal cord injury (SCI). SUMMARY OF BACKGROUND DATA Prior to clinical application of stem cell therapy for SCI, many critical issues have to be addressed including efficiency, safety, method of transplantation, and differentiation of the transplanted cells. METHODS Chronic contusive SCI was induced in 36 Sprague-Dawley rats and randomly assigned to the intralesional (IL), intravenous (IV), or control groups. At 6 weeks post injury, allogenic mesenchymal stem cells (MSCs, 1 × 10 cells) were transplanted either intralesionally or intravenously for the intervention groups. Engraftment of the transplanted MSCs was evaluated with PKH 26 staining. Differentiation was evaluated using double stain with neuronal and glial cell markers. Brain-derived neurotrophic factor and nerve growth factor (NGF) were used for neurotrophic factor expression. Basso, Beattie, and Bresnahan locomotor rating scale was used for evaluation of functional recovery. RESULTS The estimated engraftment percentage of the transplanted cells in the IL group and IV group were 36.5%, and 15.5%, respectively. The engraftment of the transplanted MSCs was higher in the IL group than in the IV group. Most of the transplanted MSCs were colocalized with GFAP in both transplantation groups. Brain-derived neurotrophic factor and NGF expression (Western blot and real-time polymerase chain reaction) in the injured spinal cord was higher in both transplanted groups compared with those in the control group. At 6 weeks post transplantation, the mean Basso, Beattie, and Bresnahan locomotor scales in the IL, IV, and control groups were 5.63 ± 0.89, 5.63 ± 1.03, and 2.88 ± 0.44, respectively. The functional recovery seen in the rats that underwent transplantation was significantly better than that in the control group (P < 0.05). CONCLUSION Although the number of engrafted cells and expression of neurotrophic factors were lower in the IV group than those in the IL group, both IL and IV transplantation of MSC in the chronic SCI gave a significant clinical improvement. However, there were no differences in differentiation of the transplanted cells between the IL group and IV group. Astrocytic differentiation of the transplanted cells was predominant. LEVEL OF EVIDENCE N/A.
Collapse
|
45
|
Nessler J, Bénardais K, Gudi V, Hoffmann A, Salinas Tejedor L, Janßen S, Prajeeth CK, Baumgärtner W, Kavelaars A, Heijnen CJ, van Velthoven C, Hansmann F, Skripuletz T, Stangel M. Effects of murine and human bone marrow-derived mesenchymal stem cells on cuprizone induced demyelination. PLoS One 2013; 8:e69795. [PMID: 23922802 PMCID: PMC3724887 DOI: 10.1371/journal.pone.0069795] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Accepted: 06/17/2013] [Indexed: 11/24/2022] Open
Abstract
For the treatment of patients with multiple sclerosis there are no regenerative approaches to enhance remyelination. Mesenchymal stem cells (MSC) have been proposed to exert such regenerative functions. Intravenous administration of human MSC reduced the clinical severity of experimental autoimmune encephalomyelitis (EAE), an animal model mimicking some aspects of multiple sclerosis. However, it is not clear if this effect was achieved by systemic immunomodulation or if there is an active neuroregeneration in the central nervous system (CNS). In order to investigate remyelination and regeneration in the CNS we analysed the effects of intravenously and intranasally applied murine and human bone marrow-derived MSC on cuprizone induced demyelination, a toxic animal model which allows analysis of remyelination without the influence of the peripheral immune system. In contrast to EAE no effects of MSC on de- and remyelination and glial cell reactions were found. In addition, neither murine nor human MSC entered the lesions in the CNS in this toxic model. In conclusion, MSC are not directed into CNS lesions in the cuprizone model where the blood-brain-barrier is intact and thus cannot provide support for regenerative processes.
Collapse
Affiliation(s)
- Jasmin Nessler
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Karelle Bénardais
- Department of Neurology, Hannover Medical School, Hannover, Germany
- Center for Systems Neuroscience, Hannover, Germany
| | - Viktoria Gudi
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Andrea Hoffmann
- Department of Trauma Surgery, Hannover Medical School, Hannover, Germany
| | - Laura Salinas Tejedor
- Department of Neurology, Hannover Medical School, Hannover, Germany
- Center for Systems Neuroscience, Hannover, Germany
| | - Stefanie Janßen
- Department of Neurology, Hannover Medical School, Hannover, Germany
- Center for Systems Neuroscience, Hannover, Germany
| | | | - Wolfgang Baumgärtner
- Center for Systems Neuroscience, Hannover, Germany
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Annemieke Kavelaars
- Department of Symptom Research, University of Texas, M.D. Anderson Cancer Center, Houston, Texas, United States of America
- Laboratory for Neuroimmunology and Developmental Origins of Disease, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Cobi J. Heijnen
- Department of Symptom Research, University of Texas, M.D. Anderson Cancer Center, Houston, Texas, United States of America
- Laboratory for Neuroimmunology and Developmental Origins of Disease, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Cindy van Velthoven
- Laboratory for Neuroimmunology and Developmental Origins of Disease, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Florian Hansmann
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
| | | | - Martin Stangel
- Department of Neurology, Hannover Medical School, Hannover, Germany
- Center for Systems Neuroscience, Hannover, Germany
| |
Collapse
|
46
|
Li J, Lepski G. Cell transplantation for spinal cord injury: a systematic review. BIOMED RESEARCH INTERNATIONAL 2013; 2013:786475. [PMID: 23484157 PMCID: PMC3581246 DOI: 10.1155/2013/786475] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Revised: 11/16/2012] [Accepted: 12/11/2012] [Indexed: 02/07/2023]
Abstract
Cell transplantation, as a therapeutic intervention for spinal cord injury (SCI), has been extensively studied by researchers in recent years. A number of different kinds of stem cells, neural progenitors, and glial cells have been tested in basic research, and most have been excluded from clinical studies because of a variety of reasons, including safety and efficacy. The signaling pathways, protein interactions, cellular behavior, and the differentiated fates of experimental cells have been studied in vitro in detail. Furthermore, the survival, proliferation, differentiation, and effects on promoting functional recovery of transplanted cells have also been examined in different animal SCI models. However, despite significant progress, a "bench to bedside" gap still exists. In this paper, we comprehensively cover publications in the field from the last years. The most commonly utilized cell lineages were covered in this paper and specific areas covered include survival of grafted cells, axonal regeneration and remyelination, sensory and motor functional recovery, and electrophysiological improvements. Finally we also review the literature on the in vivo tracking techniques for transplanted cells.
Collapse
Affiliation(s)
- Jun Li
- Department of Neurosurgery, Eberhard Karls University, 72076 Tübingen, Germany
- Department of Spine Surgery, The Affiliated Hospital of Luzhou Medical College, 646000 Luzhou, China
| | - Guilherme Lepski
- Department of Neurosurgery, Eberhard Karls University, 72076 Tübingen, Germany
- Division of Neurosurgery, Department of Neurology, Faculdade de Medicina, Universidade de São Paulo, Avnida Dr. Enéas de Carvalho Aguiar 255, 05403-000 São Paulo, SP, Brazil
| |
Collapse
|
47
|
Current world literature. Curr Opin Organ Transplant 2013; 18:111-30. [PMID: 23299306 DOI: 10.1097/mot.0b013e32835daf68] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|