1
|
Kaplan N, Kabatas S, Civelek E, Savrunlu EC, Akkoc T, Boyalı O, Öztürk E, Can H, Genc A, Karaöz E. Multiroute administration of Wharton’s jelly mesenchymal stem cells in chronic complete spinal cord injury: A phase I safety and feasibility study. World J Stem Cells 2025; 17:101675. [DOI: 10.4252/wjsc.v17.i5.101675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 01/09/2025] [Accepted: 03/27/2025] [Indexed: 05/26/2025] Open
Abstract
BACKGROUND Traumatic spinal cord injury (SCI) is a life-altering condition that results in long-term complications, including progressive neurodegeneration and cord atrophy. It presents a significant unmet medical need with extensive social and economic burdens.
AIM To evaluate the safety and preliminary efficacy of allogeneic mesenchymal stem cells derived from Wharton’s jelly (WJ-MSCs) in patients with chronic complete SCI. The primary objective was to assess whether WJ-MSCs could facilitate neurological recovery and improve the quality of life in this patient population.
METHODS This open-label, multicenter phase I study investigated the effects of administering WJ-MSCs via three delivery routes: Intrathecal (for localized spinal targeting); intramuscular (for targeting end organ); and intravenous (for systemic immunomodulation). While all three routes were used concurrently to enhance therapeutic synergy, neurological, sensory, and functional scales were used to assess overall efficacy. Participants with chronic SCI (duration of at least 6 months) who had significant impairment and disability were eligible for inclusion. WJ-MSCs were administered twice monthly for 2 months, with each route receiving a dose of 1 × 106 cells/kg. Patients were closely monitored for 1 year following treatment.
RESULTS At baseline, participants displayed considerable functional deficits, as indicated by the following scores: Functional independence measure of 77.5 ± 2.26; Modified Ashworth Scale of 15.83 ± 4.83; American Spinal Injury Association (ASIA) Motor score of 1.67 ± 2.66; ASIA Light Touch and Pin-Prick scores of 62 ± 18.42 each; Wexner Incontinence Score of 20; and Qualiveen Short Form, a validated questionnaire specifically designed to assess the impact of urinary dysfunction on quality of life in individuals with SCI, score of 32. Following WJ-MSC therapy, significant improvements were observed in all neurological functions over the 1-year follow-up. Notably, the ASIA Motor score improved significantly (χ2 = 23.938, P < 0.001), and Qualiveen Short Form scores demonstrated a substantial enhancement in quality of life (z = -2.214, P < 0.05).
CONCLUSION This phase I study, conducted without a control group, suggests that the administration of WJ-MSCs through multiple routes is both safe and potentially effective in patients with chronic complete SCI. However, the observed neurological improvements cannot be solely attributed to WJ-MSC therapy, as concurrent pharmacological and rehabilitative interventions were not controlled. These findings indicated that WJ-MSC therapy may offer a promising approach for enhancing neurological function and quality of life in this challenging patient population. Further research with larger cohorts and extended follow-up is necessary to validate these preliminary results.
Collapse
Affiliation(s)
- Necati Kaplan
- Department of Neurosurgery, Istanbul Rumeli University, Çorlu Reyap Hospital, Tekirdağ 59860, Türkiye
| | - Serdar Kabatas
- Department of Neurosurgery, University of Health Sciences Türkiye, Gaziosmanpaşa Training and Research Hospital, Istanbul 34255, Türkiye
- Center for Stem Cell & Gene Therapy Research and Practice, University of Health Sciences Türkiye, İstanbul 34255, Türkiye
| | - Erdinç Civelek
- Department of Neurosurgery, University of Health Sciences Türkiye, Gaziosmanpaşa Training and Research Hospital, Istanbul 34255, Türkiye
| | | | - Tolga Akkoc
- Tubitak Marmara Research Center, Genetic Engineering and Biotechnology Institute, Kocaeli 41470, Türkiye
| | - Osman Boyalı
- Department of Neurosurgery, University of Health Sciences Türkiye, Gaziosmanpaşa Training and Research Hospital, Istanbul 34255, Türkiye
| | - Erek Öztürk
- Department of Neurosurgery, Orthopaediezentrum Magdeburg, Magdeburg 39112, Saxony-Anhalt, Germany
| | - Halil Can
- Department of Neurosurgery, Atlas University, İstanbul 34408, Türkiye
| | - Ali Genc
- Department of Neurosurgery, Palmiye Hospital, Hatay 31200, Türkiye
| | - Erdal Karaöz
- Center for Regenerative Medicine and Stem Cell Research & Manufacturing, Liv Hospital, Istanbul 34340, Türkiye
- Department of Histology and Embryology, Istinye University, Faculty of Medicine, Zeytinburnu 34010, Istanbul, Türkiye
- Istinye University, Center for Stem Cell and Tissue Engineering Research and Practice, Beşiktaş 34340, Istanbul, Türkiye
| |
Collapse
|
2
|
Vij R, Kim H, Park H, Cheng T, Lotfi D, Chang D. Functional recovery of a 41-year-old quadriplegic spinal cord injury patient following multiple intravenous infusions of autologous adipose-derived mesenchymal stem cells: a case report. FRONTIERS IN TRANSPLANTATION 2023; 2:1287508. [PMID: 38993875 PMCID: PMC11235215 DOI: 10.3389/frtra.2023.1287508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 11/23/2023] [Indexed: 07/13/2024]
Abstract
Spinal cord injury (SCI) is a debilitating disease with clinical manifestations ranging from incomplete neurological deficits affecting sensory and motor functions to complete paralysis. Recent advancements in stem cell research have elucidated the therapeutic potential of mesenchymal stem cells (MSCs) for the treatment of patients with SCI. Here, we present a case of a 41-year-old quadriplegic male individual who experienced a traumatic C-5 incomplete SCI, after slipping off a boat in Florida Keys on August 4, 2017. He was diagnosed with C5-C6 Grade 2 anterolisthesis with flexion teardrop fracture of the anterior C6 with jumped facet on the right and perched facet on the left at C5-C6 with spinal canal stenosis. On September 12, 2019, an Individual Expanded Access Protocol was approved for administration of multiple infusions of autologous, adipose-derived MSCs (adMSCs) for the treatment of this quadriplegic incomplete C5-6 SCI patient. Thirty-four (34) recurrent infusions each with 200 million cells were administered, over a period of ∼2.5 years, which resulted in significant improvements in his quality-of-life as demonstrated by substantial improvements in SCIM-III (Spinal Cord Independence Measure III) scores. Additionally, electromyography/nerve conduction velocity (EMG/NCV) studies showed improvements in the patient's motor and sensory function. No safety concerns were presented, and no serious adverse events were reported during the entire course of treatment. Multiple intravenous infusions of autologous HB-adMSCs for treatment of SCI demonstrated significant enhancements in the patient's neurological function with improved quality-of-life. Further research is needed to evaluate the results of this study.
Collapse
Affiliation(s)
- Ridhima Vij
- Clinical Research, Hope Biosciences Research Foundation, Sugar Land, TX, United States
| | - Hosu Kim
- Cell Production, Hope Biosciences, Sugar Land, TX, United States
| | - Hyeonggeun Park
- Cell Production, Hope Biosciences, Sugar Land, TX, United States
| | - Thanh Cheng
- Clinical Research, Hope Biosciences Research Foundation, Sugar Land, TX, United States
| | - Djamchid Lotfi
- Clinical Research, Hope Biosciences Research Foundation, Sugar Land, TX, United States
| | - Donna Chang
- Clinical Research, Hope Biosciences Research Foundation, Sugar Land, TX, United States
- Cell Production, Hope Biosciences, Sugar Land, TX, United States
| |
Collapse
|
3
|
Farhana S, Kai YC, Kadir R, Sulaiman WAW, Nordin NA, Nasir NAM. The fate of adipose tissue and adipose-derived stem cells in allograft. Cell Tissue Res 2023; 394:269-292. [PMID: 37624425 DOI: 10.1007/s00441-023-03827-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 08/15/2023] [Indexed: 08/26/2023]
Abstract
Utilizing adipose tissue and adipose-derived stem cells (ADSCs) turned into a promising field of allograft in recent years. The therapeutic potential of adipose tissue and ADSCs is governed by their molecular secretions, ability to sustain multi-differentiation and self-renewal which are pivotal in reconstructive, genetic diseases, and cosmetic goals. However, revisiting the existing functional capacity of adipose tissue and ADSCs and their intricate relationship with allograft is crucial to figure out the remarkable question of safety to use in allograft due to the growing evidence of interactions between tumor microenvironment and ADSCs. For instance, the molecular secretions of adipose tissue and ADSCs induce angiogenesis, create growth factors, and control the inflammatory response; it has now been well determined. Though the existing preclinical allograft studies gave positive feedback, ADSCs and adipose tissue are attracted by some factors of tumor stroma. Moreover, allorecognition is pivotal to allograft rejection which is carried out by costimulation in a complement-dependent way and leads to the destruction of the donor cells. However, extensive preclinical trials of adipose tissue and ADSCs in allograft at molecular level are still limited. Hence, comprehensive immunomodulatory analysis could ensure the successful allograft of adipose tissue and ADSCs avoiding the oncological risk.
Collapse
Affiliation(s)
- Sadia Farhana
- Reconstructive Sciences Unit, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, 16150, Kota Bharu, Kelantan, Malaysia
| | - Yew Chun Kai
- Reconstructive Sciences Unit, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, 16150, Kota Bharu, Kelantan, Malaysia
- Hospital Universiti Sains Malaysia, 16150, Kota Bharu, Kelantan, Malaysia
| | - Ramlah Kadir
- Department of Immunology, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, 16150, Kota Bharu, Kelantan, Malaysia
| | - Wan Azman Wan Sulaiman
- Reconstructive Sciences Unit, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, 16150, Kota Bharu, Kelantan, Malaysia
- Hospital Universiti Sains Malaysia, 16150, Kota Bharu, Kelantan, Malaysia
| | - Nor Asyikin Nordin
- Department of Immunology, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, 16150, Kota Bharu, Kelantan, Malaysia
| | - Nur Azida Mohd Nasir
- Reconstructive Sciences Unit, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, 16150, Kota Bharu, Kelantan, Malaysia.
| |
Collapse
|
4
|
Gouveia D, Correia J, Cardoso A, Carvalho C, Oliveira AC, Almeida A, Gamboa Ó, Ribeiro L, Branquinho M, Sousa A, Lopes B, Sousa P, Moreira A, Coelho A, Rêma A, Alvites R, Ferreira A, Maurício AC, Martins Â. Intensive neurorehabilitation and allogeneic stem cells transplantation in canine degenerative myelopathy. Front Vet Sci 2023; 10:1192744. [PMID: 37520009 PMCID: PMC10374290 DOI: 10.3389/fvets.2023.1192744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 06/12/2023] [Indexed: 08/01/2023] Open
Abstract
Introduction Degenerative myelopathy (DM) is a neurodegenerative spinal cord disease with upper motor neurons, with progressive and chronic clinical signs, similar to amyotrophic lateral sclerosis (ALS). DM has a complex etiology mainly associated with SOD1 gene mutation and its toxic role, with no specific treatment. Daily intensive rehabilitation showed survival time near 8 months but most animals are euthanized 6-12 months after clinical signs onset. Methods This prospective controlled blinded cohort clinical study aims to evaluate the neural regeneration response ability of DM dogs subjected to an intensive neurorehabilitation protocol with mesenchymal stem cells (MSCs) transplantation. In total, 13 non-ambulatory (OFS 6 or 8) dogs with homozygous genotype DM/DM and diagnosed by exclusion were included. All were allocated to the intensive neurorehabilitation with MSCs protocol (INSCP) group (n = 8) or to the ambulatory rehabilitation protocol (ARP) group (n = 5), which differ in regard to training intensity, modalities frequency, and MSCs transplantation. The INSCP group was hospitalized for 1 month (T0 to T1), followed by MSCs transplantation (T1) and a second month (T2), whereas the ARP group was under ambulatory treatment for the same 2 months. Results Survival mean time of total population was 375 days, with 438 days for the INSCP group and 274 for the ARP group, with a marked difference on the Kaplan-Meier survival analysis. When comparing the literature's results, there was also a clear difference in the one-sample t-test (p = 0.013) with an increase in time of approximately 70%. OFS classifications between groups at each time point were significantly different (p = 0.008) by the one-way ANOVA and the independent sample t-test. Discussion This INSCP showed to be safe, feasible, and a possibility for a long progression of DM dogs with quality of life and functional improvement. This study should be continued.
Collapse
Affiliation(s)
- Débora Gouveia
- Arrábida Veterinary Hospital, Arrábida Animal Rehabilitation Center, Setubal, Portugal
- Superior School of Health, Protection and Animal Welfare, Polytechnic Institute of Lusophony, Lisboa, Portugal
- Faculty of Veterinary Medicine, Lusófona University, Lisboa, Portugal
| | - Jéssica Correia
- Arrábida Veterinary Hospital, Arrábida Animal Rehabilitation Center, Setubal, Portugal
- Faculty of Veterinary Medicine, Lusófona University, Lisboa, Portugal
| | - Ana Cardoso
- Arrábida Veterinary Hospital, Arrábida Animal Rehabilitation Center, Setubal, Portugal
- Superior School of Health, Protection and Animal Welfare, Polytechnic Institute of Lusophony, Lisboa, Portugal
| | - Carla Carvalho
- Arrábida Veterinary Hospital, Arrábida Animal Rehabilitation Center, Setubal, Portugal
| | - Ana Catarina Oliveira
- Arrábida Veterinary Hospital, Arrábida Animal Rehabilitation Center, Setubal, Portugal
- Superior School of Health, Protection and Animal Welfare, Polytechnic Institute of Lusophony, Lisboa, Portugal
| | - António Almeida
- Faculty of Veterinary Medicine, University of Lisbon, Lisboa, Portugal
| | - Óscar Gamboa
- Faculty of Veterinary Medicine, University of Lisbon, Lisboa, Portugal
| | - Lénio Ribeiro
- Faculty of Veterinary Medicine, Lusófona University, Lisboa, Portugal
| | - Mariana Branquinho
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salaza, Universidade do Porto, Porto, Portugal
- Centro de Estudos de Ciência Animal, Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto, Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Lisboa, Portugal
| | - Ana Sousa
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salaza, Universidade do Porto, Porto, Portugal
- Centro de Estudos de Ciência Animal, Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto, Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Lisboa, Portugal
| | - Bruna Lopes
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salaza, Universidade do Porto, Porto, Portugal
- Centro de Estudos de Ciência Animal, Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto, Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Lisboa, Portugal
| | - Patrícia Sousa
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salaza, Universidade do Porto, Porto, Portugal
- Centro de Estudos de Ciência Animal, Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto, Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Lisboa, Portugal
| | - Alícia Moreira
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salaza, Universidade do Porto, Porto, Portugal
- Centro de Estudos de Ciência Animal, Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto, Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Lisboa, Portugal
| | - André Coelho
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salaza, Universidade do Porto, Porto, Portugal
- Centro de Estudos de Ciência Animal, Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto, Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Lisboa, Portugal
| | - Alexandra Rêma
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salaza, Universidade do Porto, Porto, Portugal
- Centro de Estudos de Ciência Animal, Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto, Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Lisboa, Portugal
| | - Rui Alvites
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salaza, Universidade do Porto, Porto, Portugal
- Centro de Estudos de Ciência Animal, Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto, Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Lisboa, Portugal
- Instituto Universitário de Ciências da Saúde (IUCS), Cooperativa de Ensino Superior Politécnico e Universitário (CESPU), Gandra, Portugal
| | - António Ferreira
- Faculty of Veterinary Medicine, University of Lisbon, Lisboa, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Lisboa, Portugal
- CIISA - Centro Interdisciplinar-Investigáo em Saúde Animal, Faculdade de Medicina Veterinária, Av. Universi dade Técnica de Lisboa, Lisboa, Portugal
| | - Ana Colette Maurício
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salaza, Universidade do Porto, Porto, Portugal
- Centro de Estudos de Ciência Animal, Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto, Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Lisboa, Portugal
| | - Ângela Martins
- Arrábida Veterinary Hospital, Arrábida Animal Rehabilitation Center, Setubal, Portugal
- Superior School of Health, Protection and Animal Welfare, Polytechnic Institute of Lusophony, Lisboa, Portugal
- Faculty of Veterinary Medicine, Lusófona University, Lisboa, Portugal
| |
Collapse
|
5
|
Chen J, Wang L, Liu M, Gao G, Zhao W, Fu Q, Wang Y. Implantation of adipose-derived mesenchymal stem cell sheets promotes axonal regeneration and restores bladder function after spinal cord injury. Stem Cell Res Ther 2022; 13:503. [PMID: 36224621 PMCID: PMC9558366 DOI: 10.1186/s13287-022-03188-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 09/30/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cell-based therapy using adipose-derived mesenchymal stem cells (ADSCs) is a promising treatment strategy for neurogenic bladder (NB) associated with spinal cord injury (SCI). However, therapeutic efficacy is low because of inefficient cell delivery. Cell sheets improve the efficacy of cell transplantation. Therefore, this study was conducted to investigate the therapeutic efficacy of transplanting ADSC sheets into an SCI rat model and focused on the function and pathological changes of the bladder. METHODS ADSC sheets were prepared from adipose tissue of Sprague-Dawley (SD) rats using temperature-responsive cell culture dishes. Adult female SD rats were subjected to SCI by transection at the T10 level and administered ADSC sheets or gelatin sponge (the control group). Four and 8 weeks later, in vivo cystometrograms were obtained for voiding function assessment. Rats were sacrificed and the expression of various markers was analyzed in spinal and bladder tissues. RESULTS The number of β-tubulin III-positive axons in the ADSC sheet transplantation group was higher than that in the control group. Conversely, expression of glial fibrillary acidic protein in the ADSC sheet transplantation group was lower than that in the control group. Cystometry showed impairment of the voiding function after SCI, which was improved after ADSC sheet transplantation with increased high-frequency oscillation activity. Furthermore, ADSC sheet transplantation prevented disruption of the bladder urothelium in SCI rats, thereby maintaining the intact barrier. Compared with fibrosis of the bladder wall in the control group, the ADSC sheet transplantation group had normal morphology of the bladder wall and reduced tissue fibrosis as shown by downregulation of type 1 collagen. ADSC sheet transplantation also resulted in strong upregulation of contractile smooth muscle cell (SMC) markers (α-smooth muscle actin and smoothelin) and downregulation of synthetic SMC markers (MYH10 and RBP1). CONCLUSION ADSC sheet transplantation significantly improved voiding function recovery in rats after SCI. ADSC sheet transplantation is a promising cell delivery and treatment option for NB related to SCI.
Collapse
Affiliation(s)
- Jiasheng Chen
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Eastern Institute of Urologic Reconstruction, Shanghai Jiao Tong University, Shanghai, China
| | - Lin Wang
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Eastern Institute of Urologic Reconstruction, Shanghai Jiao Tong University, Shanghai, China
| | - Meng Liu
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Eastern Institute of Urologic Reconstruction, Shanghai Jiao Tong University, Shanghai, China
| | - Guo Gao
- Key Laboratory for Thin Film and Micro Fabrication of the Ministry of Education, School of Sensing Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Weixin Zhao
- Wake Forest Institute for Regenerative Medicine, Winston Salem, NC, USA
| | - Qiang Fu
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Eastern Institute of Urologic Reconstruction, Shanghai Jiao Tong University, Shanghai, China.
| | - Ying Wang
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Eastern Institute of Urologic Reconstruction, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
6
|
Kim GU, Sung SE, Kang KK, Choi JH, Lee S, Sung M, Yang SY, Kim SK, Kim YI, Lim JH, Seo MS, Lee GW. Therapeutic Potential of Mesenchymal Stem Cells (MSCs) and MSC-Derived Extracellular Vesicles for the Treatment of Spinal Cord Injury. Int J Mol Sci 2021; 22:13672. [PMID: 34948463 PMCID: PMC8703906 DOI: 10.3390/ijms222413672] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/14/2021] [Accepted: 12/18/2021] [Indexed: 12/15/2022] Open
Abstract
Spinal cord injury (SCI) is a life-threatening condition that leads to permanent disability with partial or complete loss of motor, sensory, and autonomic functions. SCI is usually caused by initial mechanical insult, followed by a cascade of several neuroinflammation and structural changes. For ameliorating the neuroinflammatory cascades, MSC has been regarded as a therapeutic agent. The animal SCI research has demonstrated that MSC can be a valuable therapeutic agent with several growth factors and cytokines that may induce anti-inflammatory and regenerative effects. However, the therapeutic efficacy of MSCs in animal SCI models is inconsistent, and the optimal method of MSCs remains debatable. Moreover, there are several limitations to developing these therapeutic agents for humans. Therefore, identifying novel agents for regenerative medicine is necessary. Extracellular vesicles are a novel source for regenerative medicine; they possess nucleic acids, functional proteins, and bioactive lipids and perform various functions, including damaged tissue repair, immune response regulation, and reduction of inflammation. MSC-derived exosomes have advantages over MSCs, including small dimensions, low immunogenicity, and no need for additional procedures for culture expansion or delivery. Certain studies have demonstrated that MSC-derived extracellular vesicles (EVs), including exosomes, exhibit outstanding chondroprotective and anti-inflammatory effects. Therefore, we reviewed the principles and patho-mechanisms and summarized the research outcomes of MSCs and MSC-derived EVs for SCI, reported to date.
Collapse
Affiliation(s)
- Gang-Un Kim
- Department of Orthopedic Surgery, Hanil General Hospital, 308 Uicheon-ro, Dobong-gu, Seoul 01450, Korea;
| | - Soo-Eun Sung
- Department of Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (S.-E.S.); (K.-K.K.); (J.-H.C.); (S.L.); (M.S.)
| | - Kyung-Ku Kang
- Department of Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (S.-E.S.); (K.-K.K.); (J.-H.C.); (S.L.); (M.S.)
| | - Joo-Hee Choi
- Department of Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (S.-E.S.); (K.-K.K.); (J.-H.C.); (S.L.); (M.S.)
| | - Sijoon Lee
- Department of Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (S.-E.S.); (K.-K.K.); (J.-H.C.); (S.L.); (M.S.)
| | - Minkyoung Sung
- Department of Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (S.-E.S.); (K.-K.K.); (J.-H.C.); (S.L.); (M.S.)
| | - Seung Yun Yang
- Department of Biomaterials Science, Life and Industry Convergence Institute, Pusan National University, Miryang 50463, Korea;
| | - Seul-Ki Kim
- Efficacy Evaluation Team, Food Science R&D Center, KolmarBNH CO., LTD, 61Heolleungro 8-gil, Seocho-gu, Seoul 06800, Korea;
| | | | - Ju-Hyeon Lim
- New Drug Development Center, Osong Medical Innovation Foundation, Chungbuk 28160, Korea;
- Department of Orthopedic Surgery, Yeungnam University College of Medicine, Yeungnam University Medical Center, 170 Hyonchung-ro, Namgu, Daegu 42415, Korea
| | - Min-Soo Seo
- Department of Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (S.-E.S.); (K.-K.K.); (J.-H.C.); (S.L.); (M.S.)
| | - Gun Woo Lee
- Cellexobio, Co. Ltd., Daegu 42415, Korea;
- Department of Orthopedic Surgery, Yeungnam University College of Medicine, Yeungnam University Medical Center, 170 Hyonchung-ro, Namgu, Daegu 42415, Korea
| |
Collapse
|
7
|
Rafiei Alavi SN, Madani Neishaboori A, Hossein H, Sarveazad A, Yousefifard M. Efficacy of adipose tissue-derived stem cells in locomotion recovery after spinal cord injury: a systematic review and meta-analysis on animal studies. Syst Rev 2021; 10:213. [PMID: 34330329 PMCID: PMC8325264 DOI: 10.1186/s13643-021-01771-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 07/21/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Considerable disparities exist on the use of adipose tissue-derived stem cells (ADSCs) for treatment of spinal cord injury (SCI). Hence, the current systematic review aimed to investigate the efficacy of ADSCs in locomotion recovery following SCI in animal models. METHODS A search was conducted in electronic databases of MEDLINE, Embase, Scopus, and Web of Science until the end of July 2019. Reference and citation tracking and searching Google and Google Scholar search engines were performed to achieve more studies. Animal studies conducted on rats having SCI which were treated with ADSCs were included in the study. Exclusion criteria were lacking a non-treated control group, not evaluating locomotion, non-rat studies, not reporting the number of transplanted cells, not reporting isolation and preparation methods of stem cells, review articles, combination therapy, use of genetically modified ADSCs, use of induced pluripotent ADSCs, and human trials. Risk of bias was assessed using Hasannejad et al.'s proposed method for quality control of SCI-animal studies. Data were analyzed in STATA 14.0 software, and based on a random effect model, pooled standardized mean difference with a 95% confidence interval was presented. RESULTS Of 588 non-duplicated papers, data from 18 articles were included. Overall risk of bias was high risk in 8 studies, some concern in 9 studies and low risk in 1 study. Current evidence demonstrated that ADSCs transplantation could improve locomotion following SCI (standardized mean difference = 1.71; 95%CI 1.29-2.13; p < 0.0001). A considerable heterogeneity was observed between the studies (I2 = 72.0%; p < 0.0001). Subgroup analysis and meta-regression revealed that most of the factors like injury model, the severity of SCI, treatment phase, injury location, and number of transplanted cells did not have a significant effect on the efficacy of ADSCs in improving locomotion following SCI (pfor odds ratios > 0.05). CONCLUSION We conclude that any number of ADSCs by any prescription routes can improve locomotion recovery in an SCI animal model, at any phase of SCI, with any severity. Given the remarkable bias about blinding, clinical translation of the present results is tough, because in addition to the complexity of the nervous system and the involvement of far more complex motor circuits in the human, blinding compliance and motor outcome assessment tests in animal studies and clinical trials are significantly different.
Collapse
Affiliation(s)
| | - Arian Madani Neishaboori
- Physiology Research Center, Iran University of Medical Sciences, Hemmat Highway, P.O Box: 14665-354, Tehran, Iran
| | - Hasti Hossein
- Physiology Research Center, Iran University of Medical Sciences, Hemmat Highway, P.O Box: 14665-354, Tehran, Iran
| | - Arash Sarveazad
- Colorectal Research Center, Iran University of Medical Sciences, Niayesh St, Satarkhan Av, P.O Box: 14665-354, 1449614535, Tehran, Iran. .,Nursing Care Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Mahmoud Yousefifard
- Physiology Research Center, Iran University of Medical Sciences, Hemmat Highway, P.O Box: 14665-354, Tehran, Iran.
| |
Collapse
|
8
|
Trends of Chitosan Based Delivery Systems in Neuroregeneration and Functional Recovery in Spinal Cord Injuries. POLYSACCHARIDES 2021. [DOI: 10.3390/polysaccharides2020031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Spinal cord injury (SCI) is one of the most complicated nervous system injuries with challenging treatment and recovery. Regenerative biomaterials such as chitosan are being reported for their wide use in filling the cavities, deliver curative drugs, and also provide adsorption sites for transplanted stem cells. Biomaterial scaffolds utilizing chitosan have shown certain therapeutic effects on spinal cord injury repair with some limitations. Chitosan-based delivery in stem cell transplantation is another strategy that has shown decent success. Stem cells can be directed to differentiate into neurons or glia in vitro. Stem cell-based therapy, biopolymer chitosan delivery strategies, and scaffold-based therapeutic strategies have been advancing as a combinatorial approach for spinal cord injury repair. In this review, we summarize the recent progress in the treatment strategies of SCI due to the use of bioactivity of chitosan-based drug delivery systems. An emphasis on the role of chitosan in neural regeneration has also been highlighted.
Collapse
|
9
|
Abstract
Traumatic spinal cord injury (SCI) results in direct and indirect damage to neural tissues, which results in motor and sensory dysfunction, dystonia, and pathological reflex that ultimately lead to paraplegia or tetraplegia. A loss of cells, axon regeneration failure, and time-sensitive pathophysiology make tissue repair difficult. Despite various medical developments, there are currently no effective regenerative treatments. Stem cell therapy is a promising treatment for SCI due to its multiple targets and reactivity benefits. The present review focuses on SCI stem cell therapy, including bone marrow mesenchymal stem cells, umbilical mesenchymal stem cells, adipose-derived mesenchymal stem cells, neural stem cells, neural progenitor cells, embryonic stem cells, induced pluripotent stem cells, and extracellular vesicles. Each cell type targets certain features of SCI pathology and shows therapeutic effects via cell replacement, nutritional support, scaffolds, and immunomodulation mechanisms. However, many preclinical studies and a growing number of clinical trials found that single-cell treatments had only limited benefits for SCI. SCI damage is multifaceted, and there is a growing consensus that a combined treatment is needed.
Collapse
Affiliation(s)
- Liyi Huang
- Department of Rehabilitation Medicine Center, 34753West China Hospital/West China School of Medicine, Sichuan University, Chengdu, Sichuan, PR China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Sichuan University, Chengdu, Sichuan Province, PR China
| | - Chenying Fu
- State Key Laboratory of Biotherapy, 34753West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Feng Xiong
- Department of Rehabilitation Medicine Center, 34753West China Hospital/West China School of Medicine, Sichuan University, Chengdu, Sichuan, PR China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Sichuan University, Chengdu, Sichuan Province, PR China
| | - Chengqi He
- Department of Rehabilitation Medicine Center, 34753West China Hospital/West China School of Medicine, Sichuan University, Chengdu, Sichuan, PR China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Sichuan University, Chengdu, Sichuan Province, PR China
| | - Quan Wei
- Department of Rehabilitation Medicine Center, 34753West China Hospital/West China School of Medicine, Sichuan University, Chengdu, Sichuan, PR China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Sichuan University, Chengdu, Sichuan Province, PR China
| |
Collapse
|
10
|
Human mesenchymal stromal/stem cells recruit resident pericytes and induce blood vessels maturation to repair experimental spinal cord injury in rats. Sci Rep 2020; 10:19604. [PMID: 33177535 PMCID: PMC7658254 DOI: 10.1038/s41598-020-76290-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 10/09/2020] [Indexed: 12/16/2022] Open
Abstract
Angiogenesis is considered to mediate the beneficial effects of mesenchymal cell therapy in spinal cord injury. After a moderate balloon-compression injury in rats, injections of either human adipose tissue-derived stromal/stem cells (hADSCs) or their conditioned culture media (CM-hADSC) elicited angiogenesis around the lesion site. Both therapies increased vascular density, but the presence of hADSCs in the tissue was required for the full maturation of new blood vessels. Only animals that received hADSC significantly improved their open field locomotion, assessed by the BBB score. Animals that received CM-hADSC only, presented haemorrhagic areas and lack pericytes. Proteomic analyses of human angiogenesis-related factors produced by hADSCs showed that both pro- and anti-angiogenic factors were produced by hADSCs in vitro, but only those related to vessel maturation were detectable in vivo. hADSCs produced PDGF-AA only after insertion into the injured spinal cord. hADSCs attracted resident pericytes expressing NG2, α-SMA, PDGF-Rβ and nestin to the lesion, potentially contributing to blood vessel maturation. We conclude that the presence of hADSCs in the injured spinal cord is essential for tissue repair.
Collapse
|
11
|
Extracellular Vesicles Derived from Epidural Fat-Mesenchymal Stem Cells Attenuate NLRP3 Inflammasome Activation and Improve Functional Recovery After Spinal Cord Injury. Neurochem Res 2020; 45:760-771. [PMID: 31953741 DOI: 10.1007/s11064-019-02950-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 11/11/2019] [Accepted: 12/28/2019] [Indexed: 12/14/2022]
Abstract
Spinal cord injury (SCI) is a devastating event which caused high mortality and morbidity. Recently, nucleotide-binding domain-like receptor protein 3 (NLRP3) inflammasome has been showed to act a critical t role in the secondly injury phase of SCI. In current study, we aimed to investigate the effect and underlying molecular mechanisms of extracellular vesicles derived from epidural fat (EF)- mesenchymal stem cells (MSCs) for the treatment of SCI. Ninety-six Sprague-Dawley rats were used for current study and randomly divided into four groups: sham group, SCI group, SCI + Saline group, SCI + Extracellular vesicles group. Basso-Beattie-Bresnahan (BBB) scores was applied to evaluate the neurological functional recovery. Cresyl violet-stained was conducted evaluate the protective effect of EF-MSCs-Extracellular vesicles on lesion volume after SCI. ELISA, immunohistochemistry assay, TUNEL assay and western blotting were conducted to investigate the underlying molecular mechanisms. Our results demonstrated that the administration of EF-MSCs-Extracellular vesicles via tail vein injection improved neurological functional recovery and reduced the lesion volume after SCI. And systemic administration of EF-MSCs-Extracellular vesicles significantly inhibited NLRP3 inflammasome activation and reduced the expression of inflammatory cytokines. Additionally, the expression levels of proapoptotic protein Bax was decreased and antiapoptotic Bcl-2 was upregulated with the treatment of EF-MSCs-Extracellular vesicles after SCI. In summary, in current study, we demonstrated for the first time that the EF-MSCs-Extracellular vesicles can improve neurological functional recovery after SCI, and the underlying molecular mechanisms may partly through the inhibition of NLRP3 inflammasome activation.
Collapse
|
12
|
Zarei-Kheirabadi M, Hesaraki M, Kiani S, Baharvand H. In vivo conversion of rat astrocytes into neuronal cells through neural stem cells in injured spinal cord with a single zinc-finger transcription factor. Stem Cell Res Ther 2019; 10:380. [PMID: 31842989 PMCID: PMC6916443 DOI: 10.1186/s13287-019-1448-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 09/23/2019] [Accepted: 10/09/2019] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Spinal cord injury (SCI) results in glial scar formation and irreversible neuronal loss, which finally leads to functional impairments and long-term disability. Our previous studies have demonstrated that the ectopic expression of Zfp521 reprograms fibroblasts and astrocytes into induced neural stem cells (iNSCs). However, it remains unclear whether treatment with Zfp521 also affects endogenous astrocytes, thus promoting further functional recovery following SCI. METHODS Rat astrocytes were transdifferentiated into neural stem cells in vitro by ZFP521 or Sox2. Then, ZFP521 was applied to the spinal cord injury site of a rat. Transduction, real-time PCR, immunohistofluorescence, and function assessments were performed at 6 weeks post-transduction to evaluate improvement and in vivo lineage reprogramming of astrocytes. RESULTS Here, we show that Zfp521 is more efficient in reprogramming cultured astrocytes compared with Sox2. In the injured spinal cord of an adult rat, resident astrocytes can be reprogrammed into neurons through a progenitor stage by Zfp521. Importantly, this treatment improves the functional abilities of the rats as evaluated by the Basso, Beattie, and Bresnahan (BBB) locomotor rating scale and further by calculation of its subscores. There was enhanced locomotor activity in the hind limbs, step length, toe spread, foot length, and paw area. In addition, motor evoked potential recordings demonstrated the functional integrity of the spinal cord. CONCLUSIONS These results have indicated that the generation of iNSCs or neurons from endogenous astrocytes by in situ reprogramming might be a potential strategy for SCI repair.
Collapse
Affiliation(s)
- Masoumeh Zarei-Kheirabadi
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, 1665659911, Iran
| | - Mahdi Hesaraki
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, 1665659911, Iran
| | - Sahar Kiani
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, 1665659911, Iran.
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, 1665659911, Iran.
- Department of Developmental Biology, University of Science and Culture, Tehran, 1461968151, Iran.
| |
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
|
Karaoz E, Tepekoy F, Yilmaz I, Subasi C, Kabatas S. Reduction of Inflammation and Enhancement of Motility after Pancreatic Islet Derived Stem Cell Transplantation Following Spinal Cord Injury. J Korean Neurosurg Soc 2019; 62:153-165. [PMID: 30840970 PMCID: PMC6411578 DOI: 10.3340/jkns.2018.0035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 06/23/2018] [Indexed: 01/01/2023] Open
Abstract
Objective Spinal cord injury (SCI) is a very serious health problem, usually caused by a trauma and accompanied by elevated levels of inflammation indicators. Stem cell-based therapy is promising some valuable strategies for its functional recovery. Nestin-positive progenitor and/or stem cells (SC) isolated from pancreatic islets (PI) show mesenchymal stem cell (MSC) characteristics. For this reason, we aimed to analyze the effects of rat pancreatic islet derived stem cell (rPI-SC) delivery on functional recovery, as well as the levels of inflammation factors following SCI.
Methods rPI-SCs were isolated, cultured and their MSC characteristics were determined through flow cytometry and immunofluorescence analysis. The experimental rat population was divided into three groups : 1) laminectomy & trauma, 2) laminectomy & trauma & phosphate-buffered saline (PBS), and 3) laminectomy+trauma+SCs. Green fluorescent protein (GFP) labelled rPI-SCs were transplanted into the injured rat spinal cord. Their motilities were evaluated with Basso, Beattie and Bresnahan (BBB) Score. After 4-weeks, spinal cord sections were analyzed for GFP labeled SCs and stained for vimentin, S100β, brain derived neurotrophic factor (BDNF), 2’,3’-cyclic-nucleotide 3'-phosphodiesterase (CNPase), vascular endothelial growth factor (VEGF) and proinflammatory (interleukin [IL]-6, transforming growth factor [TGF]-β, macrophage inflammatory protein [MIP]-2, myeloperoxidase [MPO]) and anti-inflammatory (IL-1 receptor antagonis) factors.
Results rPI-SCs were revealed to display MSC characteristics and express neural and glial cell markers including BDNF, glial fibrillary acidic protein (GFAP), fibronectin, microtubule associated protein-2a,b (MAP2a,b), β3-tubulin and nestin as well as antiinflammatory prostaglandin E2 receptor, EP3. The BBB scores showed significant motor recovery in group 3. GFP-labelled cells were localized on the injury site. In addition, decreased proinflammatory factor levels and increased intensity of anti-inflammatory factors were determined.
Conclusion Transplantation of PI-SCs might be an effective strategy to improve functional recovery following spinal cord trauma.
Collapse
Affiliation(s)
- Erdal Karaoz
- Department of Histology & Embryology, Faculty of Medicine, İstinye University, İstanbul, Turkey.,Center for Stem Cell and Tissue Engineering Research & Practice, İstinye University, İstanbul, Turkey.,Center for Regenerative Medicine and Stem Cell Research & Manufacturing (LivMedCell), İstanbul, Turkey
| | - Filiz Tepekoy
- Department of Histology & Embryology, Faculty of Medicine, İstinye University, İstanbul, Turkey
| | - Irem Yilmaz
- Center for Regenerative Medicine and Stem Cell Research & Manufacturing (LivMedCell), İstanbul, Turkey
| | - Cansu Subasi
- Center for Regenerative Medicine and Stem Cell Research & Manufacturing (LivMedCell), İstanbul, Turkey
| | - Serdar Kabatas
- Neurosurgery Clinic, Gaziosmanpasa Taksim Training and Research Hospital, İstanbul, Turkey
| |
Collapse
|
15
|
Sun X, Zhu Y, Yin HY, Guo ZY, Xu F, Xiao B, Jiang WL, Guo WM, Meng HY, Lu SB, Wang Y, Peng J. Differentiation of adipose-derived stem cells into Schwann cell-like cells through intermittent induction: potential advantage of cellular transient memory function. Stem Cell Res Ther 2018; 9:133. [PMID: 29751848 PMCID: PMC5948899 DOI: 10.1186/s13287-018-0884-3] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 04/02/2018] [Accepted: 04/20/2018] [Indexed: 12/11/2022] Open
Abstract
Background Peripheral nerve injury (PNI) is a worldwide issue associated with severe social and economic burden. Autologous nerve grafting, the gold standard treatment for peripheral nerve defects, still has a number of technical limitations. Tissue engineering technology is a novel therapeutic strategy, and mesenchymal stromal cells (MSCs) are promising seed cells for nerve tissue engineering. However, the efficiency of traditional methods for inducing the differentiation of MSCs to Schwann cell-like cells (SCLCs) remains unsatisfactory. Methods Here, we propose an intermittent induction method with alternate use of complete and incomplete induction medium to induce differentiation of adipose-derived stem cells (ASCs) to SCLCs. The time dependence of traditional induction methods and the efficiency of the intermittent induction method and traditional induction methods were evaluated and compared using immunocytochemistry, quantitative reverse transcription polymerase chain reaction (qRT-PCR), enzyme-linked immunosorbent assay (ELISA), and co-culture with the dorsal root ganglion (DRG) in vitro. Cell transplantation was used to compare the effects of the traditional induction method and the intermittent induction method in repairing sciatic nerve defects in vivo. Results The results of the present study indicated that the intermittent induction method is more efficient than traditional methods for inducing ASCs to differentiate into SCLCs. In addition, SCLCs induced by this method were closer to mature myelinating Schwann cells and were capable of secreting neurotrophins and promoting DRG axon regeneration in vitro. Furthermore, SCLCs induced by the intermittent induction method could repair sciatic nerve defects in rats by cell transplantation in vivo more effectively than those produced by traditional methods. Conclusion Intermittent induction represents a novel strategy for obtaining seed cells for use in nerve tissue engineering. Electronic supplementary material The online version of this article (10.1186/s13287-018-0884-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xun Sun
- Institute of Orthopedics, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, PLA, No.28 Fuxing Road, Beijing, 100853, People's Republic of China.,School of Medicine, Nankai University, No.94 Weijin Road, Tianjin, 300071, People's Republic of China
| | - Yun Zhu
- Institute of Orthopedics, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, PLA, No.28 Fuxing Road, Beijing, 100853, People's Republic of China.,School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, No.21 Sassoon Road, Pokfulam, 999077, Hong Kong
| | - He-Yong Yin
- Department of Surgery, Experimental Surgery and Regenerative Medicine, Ludwig-Maximilians-University (LMU), Nussbaumstr. 20, 80336, Munich, Germany
| | - Zhi-Yuan Guo
- Institute of Orthopedics, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, PLA, No.28 Fuxing Road, Beijing, 100853, People's Republic of China
| | - Feng Xu
- Institute of Orthopedics, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, PLA, No.28 Fuxing Road, Beijing, 100853, People's Republic of China
| | - Bo Xiao
- Institute of Orthopedics, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, PLA, No.28 Fuxing Road, Beijing, 100853, People's Republic of China
| | - Wen-Li Jiang
- Department of Ultrasound, Beijing Hospital, National Center of Gerontology, No.1 Dahua Road, Beijing, 100730, People's Republic of China
| | - Wei-Min Guo
- Institute of Orthopedics, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, PLA, No.28 Fuxing Road, Beijing, 100853, People's Republic of China
| | - Hao-Ye Meng
- Institute of Orthopedics, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, PLA, No.28 Fuxing Road, Beijing, 100853, People's Republic of China
| | - Shi-Bi Lu
- Institute of Orthopedics, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, PLA, No.28 Fuxing Road, Beijing, 100853, People's Republic of China
| | - Yu Wang
- Institute of Orthopedics, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, PLA, No.28 Fuxing Road, Beijing, 100853, People's Republic of China. .,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, 226007, People's Republic of China.
| | - Jiang Peng
- Institute of Orthopedics, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Lab of Musculoskeletal Trauma & War Injuries, PLA, No.28 Fuxing Road, Beijing, 100853, People's Republic of China. .,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, 226007, People's Republic of China.
| |
Collapse
|
16
|
Teixeira WGJ, Cristante AF, Marcon RM, Bispo G, Ferreira R, de Barros-Filho TEP. Granulocyte Colony-Stimulating Factor Combined with Methylprednisolone Improves Functional Outcomes in Rats with Experimental Acute Spinal Cord Injury. Clinics (Sao Paulo) 2018; 73:e235. [PMID: 29466494 PMCID: PMC5808113 DOI: 10.6061/clinics/2018/e235] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 10/17/2017] [Indexed: 12/03/2022] Open
Abstract
OBJECTIVES To evaluate the effects of combined treatment with granulocyte colony-stimulating factor (G-CSF) and methylprednisolone in rats subjected to experimental spinal cord injury. METHODS Forty Wistar rats received a moderate spinal cord injury and were divided into four groups: control (no treatment); G-CSF (G-CSF at the time of injury and daily over the next five days); methylprednisolone (methylprednisolone for 24 h); and G-CSF/Methylprednisolone (methylprednisolone for 24 h and G-CSF at the time of injury and daily over the next five days). Functional evaluation was performed using the Basso, Beattie and Bresnahan score on days 2, 7, 14, 21, 28, 35 and 42 following injury. Motor-evoked potentials were evaluated. Histological examination of the spinal cord lesion was performed immediately after euthanasia on day 42. RESULTS Eight animals were excluded (2 from each group) due to infection, a normal Basso, Beattie and Bresnahan score at their first evaluation, or autophagy, and 32 were evaluated. The combination of methylprednisolone and G-CSF promoted greater functional improvement than methylprednisolone or G-CSF alone (p<0.001). This combination also exhibited a synergistic effect, with improvements in hyperemia and cellular infiltration at the injury site (p<0.001). The groups displayed no neurophysiological differences (latency p=0.85; amplitude p=0.75). CONCLUSION Methylprednisolone plus G-CSF promotes functional and histological improvements superior to those achieved by either of these drugs alone when treating spinal cord contusion injuries in rats. Combining the two drugs did have a synergistic effect.
Collapse
Affiliation(s)
- William Gemio Jacobsen Teixeira
- Divisao de Cirurgia da Coluna, Tumores da Coluna, Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
- *Corresponding author. E-mail:
| | - Alexandre Fogaça Cristante
- Divisao de Cirurgia da Coluna, Laboratorio de Investigacao Medica, Instituto de Ortopedia e Traumatologia (IOT), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Raphael Martus Marcon
- Divisao de Cirurgia da Coluna, Laboratorio de Investigacao Medica, Instituto de Ortopedia e Traumatologia (IOT), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Gustavo Bispo
- Laboratorio de Investigacao Medica – 41 (LIM-41), Instituto de Ortopedia e Traumatologia (IOT), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Ricardo Ferreira
- Divisao de Cirurgia da Coluna, Laboratorio de Investigacao Medica, Instituto de Ortopedia e Traumatologia (IOT), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Tarcísio Eloy Pessoa de Barros-Filho
- Divisao de Cirurgia da Coluna, Laboratorio de Investigacao Medica, Instituto de Ortopedia e Traumatologia (IOT), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| |
Collapse
|