1
|
Zhang C, Morozova AY, Abakumov MA, Mel'nikov PA, Gabashvili AN, Chekhonin VP. Evaluation of the Optimal Number of Implanted Mesenchymal Stem Cells for the Treatment of Post-Traumatic Syrinx and Recovery of Motor Activity after Chronic Spinal Cord Injury. Bull Exp Biol Med 2023; 175:557-568. [PMID: 37773573 DOI: 10.1007/s10517-023-05904-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Indexed: 10/01/2023]
Abstract
The present work aims at determining the most effective dose (number) of mesenchymal stem cells (MSC) for its transplantation in order to treat chronic spinal cord injury (SCI) in mature Sprague-Dawley rats (n=24). MSC were obtained from bone marrow of 4-6-month-old Sprague-Dawley rats. Four weeks after SCI, MSC suspension (4 μl) was injected to experimental animals into the injured area in doses of 4×105, 8×105, or 106. Using MRI, diffusion tensor imaging (DTI), diffusion tensor tractography (DTT), immunohistochemistry, histological staining, and behavioral tests, we studied the effect of transplantation of MSC in different doses on the following parameters in rats with SCI: the size of lesion cavity and post-traumatic syrinx (PTS), glial scar formation, neuronal fibers remodeling, axonal regeneration and sprouting, vascularization, expression of neuronal factors, and motor functions. MSC administration improved motor function in rats after SCI due to stimulation of regeneration and sprouting of the axons, enhanced recovery of locomotor functions, reduction of PTS and the glial scar, and stimulation of vascularization and expression of the neurotrophic factors. The effects of MSC were dose-dependent; the most effective dose was 106 cells.
Collapse
Affiliation(s)
- C Zhang
- Department of Medicinal Nanobiotechnology, Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia.
- Department of Basic and Applied Neurobiology, V. P. Serbsky National Medical Research Center for Psychiatry and Narcology, Ministry of Health of the Russian Federation, Moscow, Russia.
| | - A Yu Morozova
- Department of Basic and Applied Neurobiology, V. P. Serbsky National Medical Research Center for Psychiatry and Narcology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - M A Abakumov
- Department of Medicinal Nanobiotechnology, Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia
| | - P A Mel'nikov
- Department of Medicinal Nanobiotechnology, Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia
| | - A N Gabashvili
- Department of Medicinal Nanobiotechnology, Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia
| | - V P Chekhonin
- Department of Medicinal Nanobiotechnology, Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia
- Department of Basic and Applied Neurobiology, V. P. Serbsky National Medical Research Center for Psychiatry and Narcology, Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
2
|
Sousa JPM, Stratakis E, Mano J, Marques PAAP. Anisotropic 3D scaffolds for spinal cord guided repair: Current concepts. BIOMATERIALS ADVANCES 2023; 148:213353. [PMID: 36848743 DOI: 10.1016/j.bioadv.2023.213353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/15/2023] [Accepted: 02/17/2023] [Indexed: 02/24/2023]
Abstract
A spinal cord injury (SCI) can be caused by unforeseen events such as a fall, a vehicle accident, a gunshot, or a malignant illness, which has a significant impact on the quality of life of the patient. Due to the limited regenerative potential of the central nervous system (CNS), SCI is one of the most daunting medical challenges of modern medicine. Great advances have been made in tissue engineering and regenerative medicine, which include the transition from two-dimensional (2D) to three-dimensional (3D) biomaterials. Combinatory treatments that use 3D scaffolds may significantly enhance the repair and regeneration of functional neural tissue. In an effort to mimic the chemical and physical properties of neural tissue, scientists are researching the development of the ideal scaffold made of synthetic and/or natural polymers. Moreover, in order to restore the architecture and function of neural networks, 3D scaffolds with anisotropic properties that replicate the native longitudinal orientation of spinal cord nerve fibres are being designed. In an effort to determine if scaffold anisotropy is a crucial property for neural tissue regeneration, this review focuses on the most current technological developments relevant to anisotropic scaffolds for SCI. Special consideration is given to the architectural characteristics of scaffolds containing axially oriented fibres, channels, and pores. By analysing neural cell behaviour in vitro and tissue integration and functional recovery in animal models of SCI, the therapeutic efficacy is evaluated for its successes and limitations.
Collapse
Affiliation(s)
- Joana P M Sousa
- TEMA - Centre for Mechanical Technology and Automation, Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal; LASI - Intelligent Systems Associate Laboratory, Portugal; Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas (FORTH-IESL), Heraklion, Greece; CICECO - Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, Aveiro 3810-193, Portugal
| | - Emmanuel Stratakis
- Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas (FORTH-IESL), Heraklion, Greece
| | - João Mano
- CICECO - Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, Aveiro 3810-193, Portugal
| | - Paula A A P Marques
- TEMA - Centre for Mechanical Technology and Automation, Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal; LASI - Intelligent Systems Associate Laboratory, Portugal.
| |
Collapse
|
3
|
Zhang D, Sun Y, Liu W. Motor functional recovery efficacy of scaffolds with bone marrow stem cells in rat spinal cord injury: a Bayesian network meta-analysis. Spinal Cord 2023; 61:93-98. [PMID: 35842526 DOI: 10.1038/s41393-022-00836-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 07/03/2022] [Accepted: 07/04/2022] [Indexed: 11/09/2022]
Abstract
STUDY DESIGN A Bayesian network meta-analysis. OBJECTIVE Spinal cord injury (SCI) can profoundly influence human health and has been linked to lifelong disability. More high-level evidence-based medical research is expected to evaluate the value of stem cells and biomaterial scaffold material therapy for SCI. METHODS We performed a comprehensive search of Web of Science, Cochrane databases, Embase, and PubMed databases. 18 randomized controlled trials including both scaffolds and BMSCs were included. We performed a Bayesian network meta-analysis to compare the motor functional recovery efficacy of different scaffolds with BMSCs in rat SCI. RESULTS In our Bayesian network meta-analysis, the motor functional recovery was found to benefit from scaffolds, BMSCs, and BMSCs combined with scaffolds, but the scaffold and BMSC groups had similar motor functional recovery efficacy, and the BMSCs combined with scaffolds group appeared to show better efficacy than BMSCs and scaffolds alone. Subgroup analysis showed that BMSCs+fibrin, BMSCs+ASC, BMSCs+gelatine, and BMSCs+collagen were the best four treatments for SCI in rat models. CONCLUSIONS These Bayesian network meta-analysis findings strongly indicated that BMSCs combined with scaffolds is more effective to improve motor functional recovery than BMSCs and scaffolds alone. The fibrin, gelatine, ASC, and collagen may be favourable scaffolds for the injured spinal cord and that scaffolds with BMSCs could be a promising option in regeneration therapy for patients with SCI.
Collapse
Affiliation(s)
- Dong Zhang
- Changqing District People's Hospital, Jinan, China
| | - Yifeng Sun
- Department of Orthopedic Surgery, The First Affliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, China
| | - Wei Liu
- Department of Orthopedic Surgery, The First Affliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, China.
| |
Collapse
|
4
|
Suzuki H, Imajo Y, Funaba M, Ikeda H, Nishida N, Sakai T. Current Concepts of Biomaterial Scaffolds and Regenerative Therapy for Spinal Cord Injury. Int J Mol Sci 2023; 24:ijms24032528. [PMID: 36768846 PMCID: PMC9917245 DOI: 10.3390/ijms24032528] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/05/2023] [Accepted: 01/11/2023] [Indexed: 02/03/2023] Open
Abstract
Spinal cord injury (SCI) is a catastrophic condition associated with significant neurological deficit and social and financial burdens. It is currently being managed symptomatically, with no real therapeutic strategies available. In recent years, a number of innovative regenerative strategies have emerged and have been continuously investigated in preclinical research and clinical trials. In the near future, several more are expected to come down the translational pipeline. Among ongoing and completed trials are those reporting the use of biomaterial scaffolds. The advancements in biomaterial technology, combined with stem cell therapy or other regenerative therapy, can now accelerate the progress of promising novel therapeutic strategies from bench to bedside. Various types of approaches to regeneration therapy for SCI have been combined with the use of supportive biomaterial scaffolds as a drug and cell delivery system to facilitate favorable cell-material interactions and the supportive effect of neuroprotection. In this review, we summarize some of the most recent insights of preclinical and clinical studies using biomaterial scaffolds in regenerative therapy for SCI and summarized the biomaterial strategies for treatment with simplified results data. One hundred and sixty-eight articles were selected in the present review, in which we focused on biomaterial scaffolds. We conducted our search of articles using PubMed and Medline, a medical database. We used a combination of "Spinal cord injury" and ["Biomaterial", or "Scaffold"] as search terms and searched articles published up until 30 April 2022. Successful future therapies will require these biomaterial scaffolds and other synergistic approaches to address the persistent barriers to regeneration, including glial scarring, the loss of a structural framework, and biocompatibility. This database could serve as a benchmark to progress in future clinical trials for SCI using biomaterial scaffolds.
Collapse
|
5
|
Stem Cell Strategies in Promoting Neuronal Regeneration after Spinal Cord Injury: A Systematic Review. Int J Mol Sci 2022; 23:ijms232112996. [PMID: 36361786 PMCID: PMC9657320 DOI: 10.3390/ijms232112996] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 10/09/2022] [Accepted: 10/25/2022] [Indexed: 11/25/2022] Open
Abstract
Spinal cord injury (SCI) is a devastating condition with a significant medical and socioeconomic impact. To date, no effective treatment is available that can enable neuronal regeneration and recovery of function at the damaged level. This is thought to be due to scar formation, axonal degeneration and a strong inflammatory response inducing a loss of neurons followed by a cascade of events that leads to further spinal cord damage. Many experimental studies demonstrate the therapeutic effect of stem cells in SCI due to their ability to differentiate into neuronal cells and release neurotrophic factors. Therefore, it appears to be a valid strategy to use in the field of regenerative medicine. This review aims to provide an up-to-date summary of the current research status, challenges, and future directions for stem cell therapy in SCI models, providing an overview of this constantly evolving and promising field.
Collapse
|
6
|
Yin Y, Tang L, Liu K, Ding X, Wang D, Chen L. Bone marrow mesenchymal stem cells may attenuate lipopolysaccharide-induced liver injury via inhibiting the NLRP3 inflammasome and hepatocyte pyroptosis. Curr Stem Cell Res Ther 2022; 17:361-369. [PMID: 35392791 DOI: 10.2174/1574888x17666220407103441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 11/05/2021] [Accepted: 01/12/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND The transplantation of bone marrow mesenchymal cells (BMSCs) has been shown to be an effective means of treating sepsis-related organ damage. Pytoptotic cell death, in turn, has recently been identified as a key driver of sepsis-related damage. At present, there are few studies on the effect of BMSC transplantation on pytoptotic cell death. OBJECTIVE We explored the ability of BMSCs to attenuate hepatic damage in a pyroptosis-related manner in a rat model of lipopolysaccharide (LPS)-induced liver injury. METHODS Following injury modeling and BMSC transplantation, we assessed the expression of the NLR family, pyrin domain containing 3 (NLRP3) inflammasome and key downstream pyroptosis-related signaling molecules. RESULTS It was found that BMSC transplantation was sufficient to significantly improve rat survival after LPS injection. Significantly reduced expression of the pyroptosis-related proteins NLRP3, caspase-1, IL-1β, and IL-18 in rats that had undergone BMSC transplantation compared to control animals. Notably, this activity was superior to single-agent administration of the NLRP3 inhibitor MCC950. CONCLUSION Our data suggest that BMSC transplantation may alleviate LPS-induced hepatic damage by suppressing the activation of the NLRP3 inflammasome and the induction of pyroptotic cell death.
Collapse
Affiliation(s)
- Yunyu Yin
- Department of Intensive Care Unit, The Affiliated hospital of North Sichuan Medical College, Nanchong,China
| | - Lu Tang
- Department of Intensive Care Unit, The Affiliated hospital of North Sichuan Medical College, Nanchong,China
| | - Kui Liu
- Department of Intensive Care Unit, The Affiliated hospital of North Sichuan Medical College, Nanchong,China
| | - Xuefeng Ding
- Department of Intensive Care Unit, The Affiliated hospital of North Sichuan Medical College, Nanchong,China
| | - Daqing Wang
- Department of Intensive Care Unit, The Affiliated hospital of North Sichuan Medical College, Nanchong,China
| | - Li Chen
- Department of Intensive Care Unit, The Affiliated hospital of North Sichuan Medical College, Nanchong,China
| |
Collapse
|
7
|
Li JJ, Liu H, Zhu Y, Yan L, Liu R, Wang G, Wang B, Zhao B. Animal Models for Treating Spinal Cord Injury Using Biomaterials-Based Tissue Engineering Strategies. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:79-100. [PMID: 33267667 DOI: 10.1089/ten.teb.2020.0267] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Jiao Jiao Li
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Ultimo, Australia
| | - Haifeng Liu
- Department of Orthopedics and Shanxi Medical University Second Affiliated Hospital, Taiyuan, China
| | - Yuanyuan Zhu
- Department of Pharmacy, Shanxi Medical University Second Affiliated Hospital, Taiyuan, China
| | - Lei Yan
- Department of Orthopedics and Shanxi Medical University Second Affiliated Hospital, Taiyuan, China
| | - Ruxing Liu
- Department of Orthopedics and Shanxi Medical University Second Affiliated Hospital, Taiyuan, China
| | - Guishan Wang
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China
| | - Bin Wang
- Department of Orthopedics and Shanxi Medical University Second Affiliated Hospital, Taiyuan, China
- Department of Sports Medicine and Adult Reconstruction Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Bin Zhao
- Department of Orthopedics and Shanxi Medical University Second Affiliated Hospital, Taiyuan, China
| |
Collapse
|
8
|
Lewis MJ, Jeffery ND, Olby NJ. Ambulation in Dogs With Absent Pain Perception After Acute Thoracolumbar Spinal Cord Injury. Front Vet Sci 2020; 7:560. [PMID: 33062648 PMCID: PMC7479830 DOI: 10.3389/fvets.2020.00560] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 07/14/2020] [Indexed: 12/11/2022] Open
Abstract
Acute thoracolumbar spinal cord injury (SCI) is common in dogs frequently secondary to intervertebral disc herniation. Following severe injury, some dogs never regain sensory function to the pelvic limbs or tail and are designated chronically "deep pain negative." Despite this, a subset of these dogs develop spontaneous motor recovery over time including some that recover sufficient function in their pelvic limbs to walk independently without assistance or weight support. This type of ambulation is commonly known as "spinal walking" and can take up to a year or more to develop. This review provides a comparative overview of locomotion and explores the physiology of locomotor recovery after severe SCI in dogs. We discuss the mechanisms by which post-injury plasticity and coordination between circuitry contained within the spinal cord, peripheral sensory feedback, and residual or recovered supraspinal connections might combine to underpin spinal walking. The clinical characteristics of spinal walking are outlined including what is known about the role of patient or injury features such as lesion location, timeframe post-injury, body size, and spasticity. The relationship between the emergence of spinal walking and electrodiagnostic and magnetic resonance imaging findings are also discussed. Finally, we review possible ways to predict or facilitate recovery of walking in chronically deep pain negative dogs. Improved understanding of the mechanisms of gait generation and plasticity of the surviving tissue after injury might pave the way for further treatment options and enhanced outcomes in severely injured dogs.
Collapse
Affiliation(s)
- Melissa J Lewis
- Department of Veterinary Clinical Sciences, Purdue University College of Veterinary Medicine, West Lafayette, IN, United States
| | - Nick D Jeffery
- Department of Small Animal Clinical Sciences, Texas a & M College of Veterinary Medicine and Biomedical Sciences, College Station, TX, United States
| | - Natasha J Olby
- Department of Clinical Sciences, North Carolina State University, Raleigh, NC, United States
| | | |
Collapse
|
9
|
Sadik ME, Ozturk AK, Albayar A, Branche M, Sullivan PZ, Schlosser LO, Browne KD, Jaye AH, Smith DH. A Strategy Toward Bridging a Complete Spinal Cord Lesion Using Stretch-Grown Axons. Tissue Eng Part A 2020; 26:623-635. [DOI: 10.1089/ten.tea.2019.0230] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Affiliation(s)
- Mindy Ezra Sadik
- Center for Brain Injury and Repair, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ali K. Ozturk
- Department of Neurosurgery, Pennsylvania Hospital, University of Pennsylvania Health System, Philadelphia, Pennsylvania
| | - Ahmed Albayar
- Center for Brain Injury and Repair, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Marc Branche
- Center for Brain Injury and Repair, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Patricia Zadnik Sullivan
- Department of Neurosurgery, Pennsylvania Hospital, University of Pennsylvania Health System, Philadelphia, Pennsylvania
| | - Laura O. Schlosser
- Center for Brain Injury and Repair, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kevin D. Browne
- Center for Brain Injury and Repair, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Andrew H. Jaye
- Center for Brain Injury and Repair, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Douglas H. Smith
- Center for Brain Injury and Repair, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
10
|
Yousefifard M, Nasseri Maleki S, Askarian-Amiri S, Vaccaro AR, Chapman JR, Fehlings MG, Hosseini M, Rahimi-Movaghar V. A combination of mesenchymal stem cells and scaffolds promotes motor functional recovery in spinal cord injury: a systematic review and meta-analysis. J Neurosurg Spine 2020; 32:269-284. [PMID: 31675724 DOI: 10.3171/2019.8.spine19201] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Accepted: 08/01/2019] [Indexed: 11/06/2022]
Abstract
OBJECTIVE There is controversy about the role of scaffolds as an adjunctive therapy to mesenchymal stem cell (MSC) transplantation in spinal cord injury (SCI). Thus, the authors aimed to design a meta-analysis on preclinical evidence to evaluate the effectiveness of combination therapy of scaffold + MSC transplantation in comparison with scaffolds alone and MSCs alone in improving motor dysfunction in SCI. METHODS Electronic databases including Medline, Embase, Scopus, and Web of Science were searched from inception until the end of August 2018. Two independent reviewers screened related experimental studies. Animal studies that evaluated the effectiveness of scaffolds and/or MSCs on motor function recovery following experimental SCI were included. The findings were reported as standardized mean difference (SMD) and 95% confidence interval (CI). RESULTS A total of 34 articles were included in the meta-analysis. Analyses show that combination therapy in comparison with the scaffold group alone (SMD 2.00, 95% CI 1.53-2.46, p < 0.0001), the MSCs alone (SMD 1.58, 95% CI 0.84-2.31, p < 0.0001), and the nontreated group (SMD 3.52, 95% CI 2.84-4.20, p < 0.0001) significantly improved motor function recovery. Co-administration of MSCs + scaffolds only in the acute phase of injury (during the first 3 days after injury) leads to a significant recovery compared to scaffold alone (SMD 2.18, p < 0.0001). In addition, the cotransplantation of scaffolds with bone marrow-derived MSCs (SMD 1.99, p < 0.0001) and umbilical cord-derived MSCs (SMD 1.50, p = 0.001) also improved motor function following SCI. CONCLUSIONS The findings showed that scaffolds + MSCs is more effective than scaffolds and MSCs alone in improving motor function following SCI in animal models, when used in the acute phase of injury.
Collapse
Affiliation(s)
- Mahmoud Yousefifard
- 1Physiology Research Center, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Solmaz Nasseri Maleki
- 1Physiology Research Center, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Alexander R Vaccaro
- 2Department of Orthopedics and Neurosurgery, Rothman Institute, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Jens R Chapman
- 3Swedish Neuroscience Institute, Swedish Medical Center, Seattle, Washington
| | - Michael G Fehlings
- 4Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
- 5Division of Neurosurgery, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
- 6Department of Surgery and Spine Program, University of Toronto, Ontario, Canada
| | - Mostafa Hosseini
- 7Department of Epidemiology and Biostatistics, School of Public Health, and
| | - Vafa Rahimi-Movaghar
- 8Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran; and
- 9Brain and Spinal Injuries Research Center (BASIR), Neuroscience Institute, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
Regulation of autophagy in mesenchymal stem cells modulates therapeutic effects on spinal cord injury. Brain Res 2019; 1721:146321. [PMID: 31278935 DOI: 10.1016/j.brainres.2019.146321] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 06/20/2019] [Accepted: 07/02/2019] [Indexed: 02/06/2023]
Abstract
Transplantation with mesenchymal stem cells (MSCs) has shown beneficial effects in treating spinal cord injury. Autophagy is an evolutionarily conserved process of degradation and recycling of cellular components that plays an important role in tissue homeostasis and cellular survival. Whether regulating autophagy in MSCs may affect their therapeutic potential in spinal cord injury repair has not yet been determined. In this study, autophagy was inhibited in MSCs with lentiviruses expressing short hairpin RNA (shRNA) to knock down Becn-1 expression, and autophagy was upregulated in MSCs under nutrient starvation. These MSCs were then labelled with Hoechst and applied to spinal cord-injured rats to evaluate their therapeutic effects. After transplanting MSCs into rats with spinal cord injuries, functional recovery, immunohistochemistry, and remyelination analyses were performed. After inducing autophagy, the MSCs exhibited an accumulation of LC3-positive autophagosomes in the cytoplasm. The expression levels of neurotrophic factors, including vascular endothelial growth factor and brain derived neurotrophic factor, were significantly higher in autophagic MSCs than normal MSCs. The in vivo study showed that more labelled MSCs migrated to the lesion site after induction of autophagy. Inducing autophagy in MSCs promoted functional recovery after spinal cord injury, whereas functional recovery was weak after inhibiting autophagy in MSCs. In contrast to the autophagy inhibition group, transplanting autophagic MSCs exhibited a greater positive impact on axon regeneration, growth of serotonergic fibers, blood vessel regeneration, and myelination, indicating a multifactorial contribution to spinal cord injury repair. These results suggest that autophagy plays important roles in MSCs during spinal cord injury repair. Regulation of autophagy in MSCs before in vivo transplantation may be a potential therapeutic interventional strategy for spinal cord injury.
Collapse
|
12
|
Zhang Q, Shi B, Ding J, Yan L, Thawani JP, Fu C, Chen X. Polymer scaffolds facilitate spinal cord injury repair. Acta Biomater 2019; 88:57-77. [PMID: 30710714 DOI: 10.1016/j.actbio.2019.01.056] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 01/10/2019] [Accepted: 01/28/2019] [Indexed: 12/23/2022]
Abstract
During the past decades, improving patient neurological recovery following spinal cord injury (SCI) has remained a challenge. An effective treatment for SCI would not only reduce fractured elements and isolate developing local glial scars to promote axonal regeneration but also ameliorate secondary effects, including inflammation, apoptosis, and necrosis. Three-dimensional (3D) scaffolds provide a platform in which these mechanisms can be addressed in a controlled manner. Polymer scaffolds with favorable biocompatibility and appropriate mechanical properties have been engineered to minimize cicatrization, customize drug release, and ensure an unobstructed space to promote cell growth and differentiation. These properties make polymer scaffolds an important potential therapeutic platform. This review highlights the recent developments in polymer scaffolds for SCI engineering. STATEMENT OF SIGNIFICANCE: How to improve the efficacy of neurological recovery after spinal cord injury (SCI) is always a challenge. Tissue engineering provides a promising strategy for SCI repair, and scaffolds are one of the most important elements in addition to cells and inducing factors. The review highlights recent development and future prospects in polymer scaffolds for SCI therapy. The review will guide future studies by outlining the requirements and characteristics of polymer scaffold technologies employed against SCI. Additionally, the peculiar properties of polymer materials used in the therapeutic process of SCI also have guiding significance to other tissue engineering approaches.
Collapse
|
13
|
Hasanzadeh E, Ebrahimi-Barough S, Mirzaei E, Azami M, Tavangar SM, Mahmoodi N, Basiri A, Ai J. Preparation of fibrin gel scaffolds containing MWCNT/PU nanofibers for neural tissue engineering. J Biomed Mater Res A 2019; 107:802-814. [DOI: 10.1002/jbm.a.36596] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 11/28/2018] [Accepted: 12/18/2018] [Indexed: 11/06/2022]
Affiliation(s)
- Elham Hasanzadeh
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine; Tehran University of Medical Sciences; Tehran Iran
- Department of Tissue Engineering, School of Advanced Technologies in Medicine; Mazandaran University of Medical Sciences; Sari Iran
| | - Somayeh Ebrahimi-Barough
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine; Tehran University of Medical Sciences; Tehran Iran
| | - Esmaeil Mirzaei
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies; Shiraz University of Medical Sciences; Shiraz Iran
| | - Mahmoud Azami
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine; Tehran University of Medical Sciences; Tehran Iran
| | - Seyed Mohammad Tavangar
- Department of Pathology; Shariati Hospital, Tehran University of Medical Sciences; Tehran Iran
| | - Narges Mahmoodi
- Sina Trauma and Surgery Research Center; Tehran University of Medical Sciences; Tehran Iran
| | - Arefeh Basiri
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine; Tehran University of Medical Sciences; Tehran Iran
| | - Jafar Ai
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine; Tehran University of Medical Sciences; Tehran Iran
| |
Collapse
|
14
|
Thakor DK, Wang L, Benedict D, Kabatas S, Zafonte RD, Teng YD. Establishing an Organotypic System for Investigating Multimodal Neural Repair Effects of Human Mesenchymal Stromal Stem Cells. ACTA ACUST UNITED AC 2018; 47:e58. [PMID: 30021049 DOI: 10.1002/cpsc.58] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Human mesenchymal stromal stem cells (hMSCs) hold regenerative medicine potential due to their availability, in vitro expansion readiness, and autologous feasibility. For neural repair, hMSCs show translational value in research on stroke, spinal cord injury (SCI), and traumatic brain injury. It is pivotal to establish multimodal in vitro systems to investigate molecular mechanisms underlying neural actions of hMSCs. Here, we describe a platform protocol on how to set up organotypic co-cultures of hMSCs (alone or polymer-scaffolded) with explanted adult rat dorsal root ganglia (DRGs) to determine neural injury and recovery events for designing implants to counteract neurotrauma sequelae. We emphasize in vitro hMSC propagation, polymer scaffolding, hMSC stemness maintenance, hMSC-DRG interaction profiling, and analytical formulas of neuroinflammation, trophic factor expression, DRG neurite outgrowth and tropic tracking, and in vivo verification of tailored implants in rodent models of SCI. © 2018 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Devang K Thakor
- Departments of Physical Medicine & Rehabilitation and Neurosurgery, Harvard Medical School/Spaulding Rehabilitation Hospital Network, Brigham and Women's Hospital, and Massachusetts General Hospital, Boston, Massachusetts.,Division of Spinal Cord Injury Research, VA Boston Healthcare System, Boston, Massachusetts
| | - Lei Wang
- Departments of Physical Medicine & Rehabilitation and Neurosurgery, Harvard Medical School/Spaulding Rehabilitation Hospital Network, Brigham and Women's Hospital, and Massachusetts General Hospital, Boston, Massachusetts.,Division of Spinal Cord Injury Research, VA Boston Healthcare System, Boston, Massachusetts.,Department of Neurosurgery, Wuhan Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Darcy Benedict
- Departments of Physical Medicine & Rehabilitation and Neurosurgery, Harvard Medical School/Spaulding Rehabilitation Hospital Network, Brigham and Women's Hospital, and Massachusetts General Hospital, Boston, Massachusetts.,Division of Spinal Cord Injury Research, VA Boston Healthcare System, Boston, Massachusetts
| | - Serdar Kabatas
- Departments of Physical Medicine & Rehabilitation and Neurosurgery, Harvard Medical School/Spaulding Rehabilitation Hospital Network, Brigham and Women's Hospital, and Massachusetts General Hospital, Boston, Massachusetts.,Division of Spinal Cord Injury Research, VA Boston Healthcare System, Boston, Massachusetts.,Department of Neurosurgery, Taksim Education and Teaching Hospital, University of Healthsciences, Istanbul, Turkey
| | - Ross D Zafonte
- Departments of Physical Medicine & Rehabilitation and Neurosurgery, Harvard Medical School/Spaulding Rehabilitation Hospital Network, Brigham and Women's Hospital, and Massachusetts General Hospital, Boston, Massachusetts
| | - Yang D Teng
- Departments of Physical Medicine & Rehabilitation and Neurosurgery, Harvard Medical School/Spaulding Rehabilitation Hospital Network, Brigham and Women's Hospital, and Massachusetts General Hospital, Boston, Massachusetts.,Division of Spinal Cord Injury Research, VA Boston Healthcare System, Boston, Massachusetts
| |
Collapse
|
15
|
Tian T, Yu Z, Zhang N, Chang Y, Zhang Y, Zhang L, Zhou S, Zhang C, Feng G, Huang F. Modified acellular nerve-delivering PMSCs improve functional recovery in rats after complete spinal cord transection. Biomater Sci 2018; 5:2480-2492. [PMID: 29106428 DOI: 10.1039/c7bm00485k] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Due to the poor regeneration capacity of neurons and the inhibitory microenvironment, spontaneous regeneration in spinal cord injury (SCI) remains challenging. Tissue engineering is considered a promising approach for enhancing the regeneration of SCI by reconstructing the inherent structure and improving the microenvironment. In this study, the possibility of engineering a nerve complex, which is constructed by acellular nerve delivering placenta mesenchymal stem cells (PMSCs), was assessed for the recovery of a transected spinal cord. Modified acellular nerve grafts were developed, and PMSCs labeled with green fluorescent protein (GFP) were seeded on the graft to construct the engineered nerve complex. Then, the engineered nerve complex was implanted into a 2 mm-length transected gap of the spinal cord. Four weeks after the transplantation, numerous surviving PMSCs were observed in the lesion cavity by immunofluorescence staining. Moreover, co-localization between GFP and neurofilament-200 (NF200) and Neuronal Class III β-Tubulin (Tuj1) was observed at the bridge interface. The PMSCs-graft group exhibited significant function improvement as evaluated by the Basso, Beattie and Bresnahan (BBB) locomotion score and footprint analysis. Eight weeks after surgery, the evoked response was restored in the PMSCs-graft group and numerous thick myelin sheathes were observed compared to that in the control groups. Collectively, our findings suggest that the nerve complex prepared by acellular nerve delivering PMSCs enhanced the structure and function regeneration of the spinal cord after SCI.
Collapse
Affiliation(s)
- Ting Tian
- Institute of Human Anatomy and Histology and Embryology, Otology & Neuroscience Center, Binzhou Medical University, 346 Guanhai Road, Laishan District, Shandong Province 264003, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Mochalova AE, Smirnova LA. State of the Art in the Targeted Modification of Chitosan. POLYMER SCIENCE SERIES B 2018. [DOI: 10.1134/s1560090418020045] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
17
|
Gao Y, Kong W, Li B, Ni Y, Yuan T, Guo L, Lin H, Fan H, Fan Y, Zhang X. Fabrication and characterization of collagen-based injectable and self-crosslinkable hydrogels for cell encapsulation. Colloids Surf B Biointerfaces 2018; 167:448-456. [PMID: 29709829 DOI: 10.1016/j.colsurfb.2018.04.009] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 04/02/2018] [Accepted: 04/03/2018] [Indexed: 01/01/2023]
Abstract
Injectable and self-crosslinkable hydrogels have drawn much attention for their potential application as cell delivery carriers to deliver cells to the injury site of arbitrary shape. In this study, injectable and self-crosslinkable hydrogels were designed and fabricated based on collagen type I (Col I) and activated chondroitin sulfate (CS-sNHS) by physical and chemical crosslinking without the addition of any catalysts. The physical properties of hydrogels, including mechanical properties, swelling and degradation properties, were investigated. The results demonstrated that the physical properties of hydrogels, especially the stiffness of hydrogels, were readily tuned by varying the degree of substitution (DS) of CS-sNHS without changing the concentration of collagen-based precursor. Chondrocytes were encapsulated into hydrogels to investigate the effects of hydrogels on the survival, proliferation and extracellular matrix (ECM) secretion of cells by FDA/PI staining, CCK-8 test and histological staining. The results suggested that all of these hydrogels supported the survival and ECM secretion of chondrocytes, while there was more ECM secretion around chondrocytes encapsulated in hydrogel Col I/CS-sNHS56% in which the DS of CS-sNHS was 56%. When the neutral precursor solution for hydrogel of Col I or Col I/CS-sNHS56% was subcutaneously injected into SD rats, hydrogels both displayed acceptable biocompatibility in vivo. These results imply that these injectable and self-crosslinkable hydrogels are suitable candidates for applications in the fields of cell delivery and tissue engineering.
Collapse
Affiliation(s)
- Yongli Gao
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan, 610064, China
| | - Weili Kong
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan, 610064, China
| | - Bao Li
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan, 610064, China
| | - Yilu Ni
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan, 610064, China
| | - Tun Yuan
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan, 610064, China
| | - Likun Guo
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan, 610064, China.
| | - Hai Lin
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan, 610064, China
| | - Hongsong Fan
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan, 610064, China
| | - Yujiang Fan
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan, 610064, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, Sichuan, 610064, China
| |
Collapse
|
18
|
Multichannel polymer scaffold seeded with activated Schwann cells and bone mesenchymal stem cells improves axonal regeneration and functional recovery after rat spinal cord injury. Acta Pharmacol Sin 2017; 38:623-637. [PMID: 28392569 DOI: 10.1038/aps.2017.11] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 01/23/2017] [Indexed: 12/17/2022]
Abstract
The adult mammalian CNS has a limited capacity to regenerate after traumatic injury. In this study, a combinatorial strategy to promote axonal regeneration and functional recovery after spinal cord injury (SCI) was evaluated in adult rats. The rats were subjected to a complete transection in the thoracic spinal cord, and multichannel scaffolds seeded with activated Schwann cells (ASCs) and/or rat bone marrow-derived mesenchymal stem cells (MSCs) were acutely grafted into the 3-mm-wide transection gap. At 4 weeks post-transplantation and thereafter, the rats receiving scaffolds seeded with ASCs and MSCs exhibited significant recovery of nerve function as shown by the Basso, Beattie and Bresnahan (BBB) score and electrophysiological test results. Immunohistochemical analyses at 4 and 8 weeks after transplantation revealed that the implanted MSCs at the lesion/graft site survived and differentiated into neuron-like cells and co-localized with host neurons. Robust bundles of regenerated fibers were identified in the lesion/graft site in the ASC and MSC co-transplantation rats, and neurofilament 200 (NF) staining confirmed that these fibers were axons. Furthermore, myelin basic protein (MBP)-positive myelin sheaths were also identified at the lesion/graft site and confirmed via electron microscopy. In addition to expressing mature neuronal markers, sparse MSC-derived neuron-like cells expressed choline acetyltransferase (ChAT) at the injury site of the ASC and MSC co-transplantation rats. These findings suggest that co-transplantation of ASCs and MSCs in a multichannel polymer scaffold may represent a novel combinatorial strategy for the treatment of spinal cord injury.
Collapse
|
19
|
Increased Understanding of Stem Cell Behavior in Neurodegenerative and Neuromuscular Disorders by Use of Noninvasive Cell Imaging. Stem Cells Int 2016; 2016:6235687. [PMID: 26997958 PMCID: PMC4779824 DOI: 10.1155/2016/6235687] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 01/07/2016] [Accepted: 01/11/2016] [Indexed: 12/13/2022] Open
Abstract
Numerous neurodegenerative and neuromuscular disorders are associated with cell-specific depletion in the human body. This imbalance in tissue homeostasis is in healthy individuals repaired by the presence of endogenous stem cells that can replace the lost cell type. However, in most disorders, a genetic origin or limited presence or exhaustion of stem cells impairs correct cell replacement. During the last 30 years, methods to readily isolate and expand stem cells have been developed and this resulted in a major change in the regenerative medicine field as it generates sufficient amount of cells for human transplantation applications. Furthermore, stem cells have been shown to release cytokines with beneficial effects for several diseases. At present however, clinical stem cell transplantations studies are struggling to demonstrate clinical efficacy despite promising preclinical results. Therefore, to allow stem cell therapy to achieve its full potential, more insight in their in vivo behavior has to be achieved. Different methods to noninvasively monitor these cells have been developed and are discussed. In some cases, stem cell monitoring even reached the clinical setting. We anticipate that by further exploring these imaging possibilities and unraveling their in vivo behavior further improvement in stem cell transplantations will be achieved.
Collapse
|
20
|
Morris R, Whishaw IQ. A Proposal for a Rat Model of Spinal Cord Injury Featuring the Rubrospinal Tract and its Contributions to Locomotion and Skilled Hand Movement. Front Neurosci 2016; 10:5. [PMID: 26858587 PMCID: PMC4728831 DOI: 10.3389/fnins.2016.00005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 01/07/2016] [Indexed: 11/21/2022] Open
Abstract
Spinal cord injury and repair is a dynamic field of research. The development of reliable animal models of traumatic spinal cord injury has been invaluable in providing a wealth of information regarding the pathological consequences and recovery potential of this condition. A number of injury models have been instrumental in the elaboration and the validation of therapeutic interventions aimed at reversing this once thought permanent condition. In general, the study of spinal cord injury and repair is made difficult by both its anatomical complexity and the complexity of the behavior it mediates. In this perspective paper, we suggest a new model for spinal cord investigation that simplifies problems related to both the functional and anatomical complexity of the spinal cord. We begin by reviewing and contrasting some of the most common animal models used for investigating spinal cord dysfunction. We then consider two widely used models of spinal deficit-recovery, one involving the corticospinal tracts (CTS) and the other the rubrospinal tract (RST). We argue that the simplicity of the function of the RST makes it a useful model for studying the cord and its functional repair. We also reflect on two obstacles that have hindered progress in the pre-clinical field, delaying translation to the clinical setup. The first is recovery of function without reconnection of the transected descending fibers and the second is the use of behavioral paradigms that are not under the control of the descending fiber pathway under scrutiny.
Collapse
Affiliation(s)
- Renée Morris
- Translational Neuroscience Facility, School of Medical Sciences, The University of New South Wales Australia Sydney, NSW, Australia
| | - Ian Q Whishaw
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience, University of Lethbridge Lethbridge, AB, Canada
| |
Collapse
|
21
|
A. Elawady M, M. Elmaghrabi M, Ebrahim N, A. Elawady M, Sabry D, Shamaa A, Ragaei A. Therapeutic Potential of Bone Marrow Derived Mesenchymal Stem Cells in Modulating Astroglyosis of Surgical Induced Experimental Spinal Cord Injury. ACTA ACUST UNITED AC 2016. [DOI: 10.4236/abb.2016.76024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
22
|
Pawar K, Cummings BJ, Thomas A, Shea LD, Levine A, Pfaff S, Anderson AJ. Biomaterial bridges enable regeneration and re-entry of corticospinal tract axons into the caudal spinal cord after SCI: Association with recovery of forelimb function. Biomaterials 2015; 65:1-12. [PMID: 26134079 PMCID: PMC4523232 DOI: 10.1016/j.biomaterials.2015.05.032] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 05/18/2015] [Indexed: 12/16/2022]
Abstract
Severed axon tracts fail to exhibit robust or spontaneous regeneration after spinal cord injury (SCI). Regeneration failure reflects a combination of factors, including the growth state of neuronal cell bodies and the regeneration-inhibitory environment of the central nervous system. However, while spared circuitry can be retrained, target reinnervation depends on longitudinally directed regeneration of transected axons. This study describes a biodegradable implant using poly(lactide-co-glycolide) (PLG) bridges as a carrier scaffold to support regeneration after injury. In order to detect regeneration of descending neuronal tracts into the bridge, and beyond into intact caudal parenchyma, we developed a mouse cervical implantation model and employed Crym:GFP transgenic mice. Characterization of Crym:GFP mice revealed that descending tracts, including the corticospinal tract, were labeled by green fluorescent protein (GFP), while ascending sensory neurons and fibers were not. Robust co-localization between GFP and neurofilament-200 (NF-200) as well as GFP and GAP-43 was observed at both the rostral and caudal bridge/tissue interface. No evidence of similar regeneration was observed in mice that received gelfoam at the lesion site as controls. Minimal co-localization between GFP reporter labeling and macrophage markers was observed. Taken together, these data suggest that axons originating from descending fiber tracts regenerated, entered into the PLG bridge at the rostral margin, continued through the bridge site, and exited to re-enter host tissue at the caudal edge of the intact bridge. Finally, regeneration through implanted bridges was associated with a reduction in ipsilateral forelimb errors on a horizontal ladder task.
Collapse
Affiliation(s)
- Kiran Pawar
- Institute for Memory Impairments and Neurological Disorders (iMIND), University of California, Irvine, CA, USA
| | - Brian J Cummings
- Institute for Memory Impairments and Neurological Disorders (iMIND), University of California, Irvine, CA, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, USA; Department of Physical Medicine and Rehabilitation, University of California, Irvine, CA, USA; Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA
| | - Aline Thomas
- Department of Chemical and Biological Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL, USA
| | - Lonnie D Shea
- Department of Chemical and Biological Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL, USA
| | | | | | - Aileen J Anderson
- Institute for Memory Impairments and Neurological Disorders (iMIND), University of California, Irvine, CA, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, USA; Department of Physical Medicine and Rehabilitation, University of California, Irvine, CA, USA; Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA.
| |
Collapse
|
23
|
Auletta JJ, Eid SK, Wuttisarnwattana P, Silva I, Metheny L, Keller MD, Guardia-Wolff R, Liu C, Wang F, Bowen T, Lee Z, Solchaga LA, Ganguly S, Tyler M, Wilson DL, Cooke KR. Human mesenchymal stromal cells attenuate graft-versus-host disease and maintain graft-versus-leukemia activity following experimental allogeneic bone marrow transplantation. Stem Cells 2015; 33:601-14. [PMID: 25336340 DOI: 10.1002/stem.1867] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 09/08/2014] [Accepted: 09/29/2014] [Indexed: 12/22/2022]
Abstract
We sought to define the effects and underlying mechanisms of human, marrow-derived mesenchymal stromal cells (hMSCs) on graft-versus-host disease (GvHD) and graft-versus-leukemia (GvL) activity. Irradiated B6D2F1 mice given C57BL/6 BM and splenic T cells and treated with hMSCs had reduced systemic GvHD, donor T-cell expansion, and serum TNFα and IFNγ levels. Bioluminescence imaging demonstrated that hMSCs redistributed from lungs to abdominal organs within 72 hours, and target tissues harvested from hMSC-treated allogeneic BMT (alloBMT) mice had less GvHD than untreated controls. Cryoimaging more precisely revealed that hMSCs preferentially distributed to splenic marginal zones and regulated T-cell expansion in the white pulp. Importantly, hMSCs had no effect on in vitro cytotoxic T-cell activity and preserved potent GvL effects in vivo. Mixed leukocyte cultures containing hMSCs exhibited decreased T-cell proliferation, reduced TNFα, IFNγ, and IL-10 but increased PGE2 levels. Indomethacin and E-prostanoid 2 (EP2) receptor antagonisms both reversed while EP2 agonism restored hMSC-mediated in vitro T-cell suppression, confirming the role for PGE2 . Furthermore, cyclo-oxygenase inhibition following alloBMT abrogated the protective effects of hMSCs. Together, our data show that hMSCs preserve GvL activity and attenuate GvHD and reveal that hMSC biodistribute to secondary lymphoid organs wherein they attenuate alloreactive T-cell proliferation likely through PGE2 induction.
Collapse
Affiliation(s)
- Jeffery J Auletta
- Host Defense Program, Hematology/Oncology/BMT and Infectious Diseases, Nationwide Children's Hospital, Columbus, Ohio, USA; Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Gładysz D, Hozyasz KK. Stem cell regenerative therapy in alveolar cleft reconstruction. Arch Oral Biol 2015; 60:1517-32. [PMID: 26263541 DOI: 10.1016/j.archoralbio.2015.07.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 05/23/2015] [Accepted: 07/04/2015] [Indexed: 12/17/2022]
Abstract
Achieving a successful and well-functioning reconstruction of craniofacial deformities still remains a challenge. As for now, autologous bone grafting remains the gold standard for alveolar cleft reconstruction. However, its aesthetic and functional results often remain unsatisfactory, which carries a long-term psychosocial and medical sequelae. Therefore, searching for novel therapeutic approaches is strongly indicated. With the recent advances in stem cell research, cell-based tissue engineering strategies move from the bench to the patients' bedside. Successful stem cell engineering employs a carefully selected stem cell source, a biodegradable scaffold with osteoconductive and osteoinductive properties, as well as an addition of growth factors or cytokines to enhance osteogenesis. This review highlights recent advances in mesenchymal stem cell tissue engineering, discusses animal models and case reports of stem cell enhanced bone regeneration, as well as ongoing clinical trials.
Collapse
Affiliation(s)
- Dominika Gładysz
- Department of Pediatrics, Institute of Mother and Child, Warsaw, Poland
| | - Kamil K Hozyasz
- Department of Pediatrics, Institute of Mother and Child, Warsaw, Poland.
| |
Collapse
|
25
|
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
|
26
|
Complete rat spinal cord transection as a faithful model of spinal cord injury for translational cell transplantation. Sci Rep 2015; 5:9640. [PMID: 25860664 PMCID: PMC5381701 DOI: 10.1038/srep09640] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 03/09/2015] [Indexed: 01/09/2023] Open
Abstract
Spinal cord injury (SCI) results in neural loss and consequently motor and sensory impairment below the injury. There are currently no effective therapies for the treatment of traumatic SCI in humans. Various animal models have been developed to mimic human SCI. Widely used animal models of SCI are complete or partial transection or experimental contusion and compression, with both bearing controversy as to which one more appropriately reproduces the human SCI functional consequences. Here we present in details the widely used procedure of complete spinal cord transection as a faithful animal model to investigate neural and functional repair of the damaged tissue by exogenous human transplanted cells. This injury model offers the advantage of complete damage to a spinal cord at a defined place and time, is relatively simple to standardize and is highly reproducible.
Collapse
|
27
|
Abstract
ABSTRACT Restoration of lost neuronal function after spinal cord injury still remains a considerable challenge for current medicine. Over the last decade, regenerative medicine has recorded rapid and promising advancements in stem cell research, genetic engineering and the progression of new sophisticated biomaterials as well as nanotechnology. This advancement has also been reflected in neural tissue engineering, where, along with the development of a new generation of well-designed biopolymer scaffolds, multifactorial therapeutic strategies are being validated in order to determine the greatest possible repair efficacy of the complex CNS pathophysiology. Much attention is currently focused on the designing of multifunctional polymer scaffolds as systems for targeted drug or gene delivery, electrical stimulation or as substrates creating a special micro-environment, promoting the growth and desired differentiation of various cell lines. In this review, the latest advances in biomaterial technology together with various combinatorial strategies designed to treat spinal cord injury treatment are summarized and discussed.
Collapse
|
28
|
Shim SW, Kwon DY, Lee BN, Kwon JS, Park JH, Lee JH, Kim JH, Lee IW, Shin JW, Lee HB, Kim WD, Kim MS. Evaluation of small intestine submucosa and poly(caprolactone-co-lactide) conduits for peripheral nerve regeneration. Tissue Eng Part A 2015; 21:1142-51. [PMID: 25435200 PMCID: PMC4356220 DOI: 10.1089/ten.tea.2014.0165] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 10/30/2014] [Indexed: 01/26/2023] Open
Abstract
The present study employed nerve guidance conduits (NGCs) only, which were made of small intestine submucosa (SIS) and poly(caprolactone-co-lactide) (PCLA) to promote nerve regeneration in a peripheral nerve injury (PNI) model with nerve defects of 15 mm. The SIS- and PCLA-NGCs were easily prepared by rolling of a SIS sheet and a bioplotter using PCLA, respectively. The prepared SIS- and PCLA-NGCs fulfilled the general requirement for use as artificial peripheral NGCs such as easy fabrication, reproducibility for mass production, suturability, sterilizability, wettability, and proper mechanical properties to resist collapsing when applied to in vivo implantation. The SIS- and PCLA-NGCs appeared to be well integrated into the host sciatic nerve without causing dislocations and serious inflammation. All NGCs stably maintained their NGC shape for 8 weeks without collapsing, which matched well with the nerve regeneration rate. Staining of the NGCs in the longitudinal direction showed that the regenerated nerves grew successfully from the SIS- and PCLA-NGCs through the sciatic nerve-injured gap and connected from the proximal to distal direction along the NGC axis. SIS-NGCs exhibited a higher nerve regeneration rate than PCLA-NGCs. Collectively, our results indicate that SIS- and PCLA-NGCs induced nerve regeneration in a PNI model, a finding that has significant implications in the future with regard to the feasibility of clinical nerve regeneration with SIS- and PCLA-NGCs prepared through an easy fabrication method using promising biomaterials.
Collapse
Affiliation(s)
- Sun Woo Shim
- Department of Molecular Science and Technology, Ajou University, Suwon, Korea
| | - Doo Yeon Kwon
- Department of Molecular Science and Technology, Ajou University, Suwon, Korea
| | - Bit Na Lee
- Department of Molecular Science and Technology, Ajou University, Suwon, Korea
| | - Jin Seon Kwon
- Department of Molecular Science and Technology, Ajou University, Suwon, Korea
| | - Ji Hoon Park
- Department of Molecular Science and Technology, Ajou University, Suwon, Korea
| | - Jun Hee Lee
- Nature-Inspired Mechanical System Team, Korea Institute of Machinery and Materials, Daejeon, Korea
| | - Jae Ho Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, Korea
| | - Il Woo Lee
- Department of Neurosurgery, Catholic University of Korea, Daejeon, Korea
| | - Jung-Woog Shin
- Department of Biomedical Engineering, Inje University, Gimhae, Korea
| | - Hai Bang Lee
- Department of Molecular Science and Technology, Ajou University, Suwon, Korea
| | - Wan-Doo Kim
- Nature-Inspired Mechanical System Team, Korea Institute of Machinery and Materials, Daejeon, Korea
| | - Moon Suk Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, Korea
| |
Collapse
|
29
|
Mead B, Berry M, Logan A, Scott RAH, Leadbeater W, Scheven BA. Stem cell treatment of degenerative eye disease. Stem Cell Res 2015; 14:243-57. [PMID: 25752437 PMCID: PMC4434205 DOI: 10.1016/j.scr.2015.02.003] [Citation(s) in RCA: 156] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 02/12/2015] [Accepted: 02/14/2015] [Indexed: 12/16/2022] Open
Abstract
Stem cell therapies are being explored extensively as treatments for degenerative eye disease, either for replacing lost neurons, restoring neural circuits or, based on more recent evidence, as paracrine-mediated therapies in which stem cell-derived trophic factors protect compromised endogenous retinal neurons from death and induce the growth of new connections. Retinal progenitor phenotypes induced from embryonic stem cells/induced pluripotent stem cells (ESCs/iPSCs) and endogenous retinal stem cells may replace lost photoreceptors and retinal pigment epithelial (RPE) cells and restore vision in the diseased eye, whereas treatment of injured retinal ganglion cells (RGCs) has so far been reliant on mesenchymal stem cells (MSC). Here, we review the properties of non-retinal-derived adult stem cells, in particular neural stem cells (NSCs), MSC derived from bone marrow (BMSC), adipose tissues (ADSC) and dental pulp (DPSC), together with ESC/iPSC and discuss and compare their potential advantages as therapies designed to provide trophic support, repair and replacement of retinal neurons, RPE and glia in degenerative retinal diseases. We conclude that ESCs/iPSCs have the potential to replace lost retinal cells, whereas MSC may be a useful source of paracrine factors that protect RGC and stimulate regeneration of their axons in the optic nerve in degenerate eye disease. NSC may have potential as both a source of replacement cells and also as mediators of paracrine treatment.
Collapse
Affiliation(s)
- Ben Mead
- Neurotrauma Research Group, Neurobiology Section, School of Clinical and Experimental Medicine, University of Birmingham, B15 2TT, UK; School of Dentistry, University of Birmingham, B4 6NN, UK.
| | - Martin Berry
- Neurotrauma Research Group, Neurobiology Section, School of Clinical and Experimental Medicine, University of Birmingham, B15 2TT, UK
| | - Ann Logan
- Neurotrauma Research Group, Neurobiology Section, School of Clinical and Experimental Medicine, University of Birmingham, B15 2TT, UK
| | - Robert A H Scott
- Neurotrauma Research Group, Neurobiology Section, School of Clinical and Experimental Medicine, University of Birmingham, B15 2TT, UK
| | - Wendy Leadbeater
- Neurotrauma Research Group, Neurobiology Section, School of Clinical and Experimental Medicine, University of Birmingham, B15 2TT, UK
| | - Ben A Scheven
- School of Dentistry, University of Birmingham, B4 6NN, UK
| |
Collapse
|
30
|
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
|
31
|
Thermo-Responsive Injectable MPEG-Polyester Diblock Copolymers for Sustained Drug Release. Polymers (Basel) 2014. [DOI: 10.3390/polym6102670] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
|
32
|
Abstract
Spinal cord injury is a complex pathology often resulting in functional impairment and paralysis. Gene therapy has emerged as a possible solution to the problems of limited neural tissue regeneration through the administration of factors promoting axonal growth, while also offering long-term local delivery of therapeutic molecules at the injury site. Of note, gene therapy is our response to the requirements of neural and glial cells following spinal cord injury, providing, in a time-dependent manner, growth substances for axonal regeneration and eliminating axonal growth inhibitors. Herein, we explore different gene therapy strategies, including targeting gene expression to modulate the presence of neurotrophic growth or survival factors and increase neural tissue plasticity. Special attention is given to describing advances in viral and non-viral gene delivery systems, as well as the available routes of gene delivery. Finally, we discuss the future of combinatorial gene therapies and give consideration to the implementation of gene therapy in humans.
Collapse
|
33
|
Kim MS, Lee HB. Perspectives on tissue-engineered nerve regeneration for the treatment of spinal cord injury. Tissue Eng Part A 2014; 20:1781-3. [PMID: 24568624 PMCID: PMC4085818 DOI: 10.1089/ten.tea.2014.0094] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 02/14/2014] [Indexed: 11/12/2022] Open
Abstract
Over the past few decades, substantial progress has been made to safely improve nerve function in spinal cord injury (SCI) patients through the regeneration of injured nerve tissue. This perspective focuses on an extensive overview of SCI research as well as tissue-engineered nerve regeneration for the treatment of SCI.
Collapse
Affiliation(s)
- Moon Suk Kim
- Department of Molecular Science and Technology, Ajou University , Suwon, Korea
| | | |
Collapse
|
34
|
Choi JW, Park JK, Chang JW, Kim DY, Kim MS, Shin YS, Kim CH. Small intestine submucosa and mesenchymal stem cells composite gel for scarless vocal fold regeneration. Biomaterials 2014; 35:4911-8. [DOI: 10.1016/j.biomaterials.2014.03.008] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 03/03/2014] [Indexed: 12/09/2022]
|
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
|
Granger N, Franklin RJM, Jeffery ND. Cell therapy for spinal cord injuries: what is really going on? Neuroscientist 2014; 20:623-38. [PMID: 24415275 DOI: 10.1177/1073858413514635] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
During the last two decades, many experiments have examined the ability of cell transplants to ameliorate the loss of function after spinal cord injuries, with the hope of developing interventions to benefit patients. Although many reports suggest positive effects, there is growing concern over the quality of the available preclinical data. It is therefore important to ask whether this worldwide investigative process is close to defining a cell transplant protocol that could be translated into human patients with a realistic chance of success. This review systematically examines the strength of the preclinical evidence and outlines mechanisms by which transplanted cells may mediate their effects in spinal cord injuries. First, we examined changes in voluntary movements in the forelimb associated with cell transplants after partial cervical lesions. Second, we examined the efficacy of transplanted cells to restore electrophysiological conduction across a complete thoracic lesion. We postulated that cell therapies found to be successful in both models could reasonably have potential to treat human patients. We conclude that although there are data to support a beneficial effect of cell transplantation, most reports provide only weak evidence because of deficits in experimental design. The mechanisms by which transplanted cells mediate their functional effects remain unclear.
Collapse
Affiliation(s)
- Nicolas Granger
- School of Veterinary Sciences, University of Bristol, Bristol, UK
| | - Robin J M Franklin
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Nick D Jeffery
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
| |
Collapse
|
37
|
Panta P, Kim DY, Kwon JS, Son AR, Lee KW, Kim MS. Protein Drug-Loaded Polymeric Nanoparticles. ACTA ACUST UNITED AC 2014. [DOI: 10.4236/jbise.2014.710082] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
38
|
Management strategies for acute spinal cord injury: current options and future perspectives. Curr Opin Crit Care 2013; 18:651-60. [PMID: 23104069 DOI: 10.1097/mcc.0b013e32835a0e54] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
PURPOSE OF REVIEW Spinal cord injury is a devastating acute neurological condition with loss of function and poor long-term prognosis. This review summarizes current management strategies and innovative concepts on the horizon. RECENT FINDINGS The routine use of steroids in patients with spinal cord injuries has been largely abandoned and considered a 'harmful standard of care'. Prospective trials have shown that early spine stabilization within 24 h results in decreased secondary complication rates. Neuronal plasticity and axonal regeneration in the adult spinal cord are limited due to myelin-associated inhibitory molecules, such as Nogo-A. The experimental inhibition of Nogo-A ameliorates axonal sprouting and functional recovery in animal models. SUMMARY General management strategies for acute spinal cord injury consist of protection of airway, breathing, oxygenation and control of blood loss with maintenance of blood pressure. Unstable spine fractures should be stabilized early to allow unrestricted mobilization of patients with spinal cord injuries and to decrease preventable complications. Steroids are largely considered obsolete and have been abandoned in clinical guidelines. Nogo-A represents a promising new pharmacological target to promote sprouting of injured axons and restore function. Prospective clinical trials of Nogo-A inhibition in patients with spinal cord injuries are currently under way.
Collapse
|
39
|
Volpato FZ, Führmann T, Migliaresi C, Hutmacher DW, Dalton PD. Using extracellular matrix for regenerative medicine in the spinal cord. Biomaterials 2013; 34:4945-55. [PMID: 23597407 DOI: 10.1016/j.biomaterials.2013.03.057] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Accepted: 03/20/2013] [Indexed: 12/12/2022]
Abstract
Regeneration within the mammalian central nervous system (CNS) is limited, and traumatic injury often leads to permanent functional motor and sensory loss. The lack of regeneration following spinal cord injury (SCI) is mainly caused by the presence of glial scarring, cystic cavitation and a hostile environment to axonal growth at the lesion site. The more prominent experimental treatment strategies focus mainly on drug and cell therapies, however recent interest in biomaterial-based strategies are increasing in number and breadth. Outside the spinal cord, approaches that utilize the extracellular matrix (ECM) to promote tissue repair show tremendous potential for various application including vascular, skin, bone, cartilage, liver, lung, heart and peripheral nerve tissue engineering (TE). Experimentally, it is unknown if these approaches can be successfully translated to the CNS, either alone or in combination with synthetic biomaterial scaffolds. In this review we outline the first attempts to apply the potential of ECM-based biomaterials and combining cell-derived ECM with synthetic scaffolds.
Collapse
Affiliation(s)
- Fabio Zomer Volpato
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove 4059, Australia
| | | | | | | | | |
Collapse
|
40
|
Perán M, García MA, Lopez-Ruiz E, Jiménez G, Marchal JA. How Can Nanotechnology Help to Repair the Body? Advances in Cardiac, Skin, Bone, Cartilage and Nerve Tissue Regeneration. MATERIALS 2013; 6:1333-1359. [PMID: 28809213 PMCID: PMC5452318 DOI: 10.3390/ma6041333] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2013] [Revised: 03/20/2013] [Accepted: 03/20/2013] [Indexed: 12/15/2022]
Abstract
Nanotechnologists have become involved in regenerative medicine via creation of biomaterials and nanostructures with potential clinical implications. Their aim is to develop systems that can mimic, reinforce or even create in vivo tissue repair strategies. In fact, in the last decade, important advances in the field of tissue engineering, cell therapy and cell delivery have already been achieved. In this review, we will delve into the latest research advances and discuss whether cell and/or tissue repair devices are a possibility. Focusing on the application of nanotechnology in tissue engineering research, this review highlights recent advances in the application of nano-engineered scaffolds designed to replace or restore the followed tissues: (i) skin; (ii) cartilage; (iii) bone; (iv) nerve; and (v) cardiac.
Collapse
Affiliation(s)
- Macarena Perán
- Department of Health Sciences, University of Jaén, Campus Las Lagunillas, S/N, Jaén 23071, Spain.
| | - María Angel García
- Research Unit, University Hospital "Virgen de las Nieves", Avda. de las Fuerzas Armadas, 2, Granada 18014, Spain.
| | - Elena Lopez-Ruiz
- Department of Health Sciences, University of Jaén, Campus Las Lagunillas, S/N, Jaén 23071, Spain.
| | - Gema Jiménez
- Biopathology and Regenerative Medicine Institute (IBIMER), University of Granada, Avda. del Conocimiento S/N. CP Armilla, Granada 18100, Spain.
| | - Juan Antonio Marchal
- Biopathology and Regenerative Medicine Institute (IBIMER), University of Granada, Avda. del Conocimiento S/N. CP Armilla, Granada 18100, Spain.
- Department of Human Anatomy and Embryology, University of Granada, Avda. De Madrid, 11, Granada 18012, Spain.
| |
Collapse
|
41
|
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
|
42
|
Wang M, Chen X, Schreyer DJ. Spinal Cord Repair by Means of Tissue Engineered Scaffolds. EMERGING TRENDS IN CELL AND GENE THERAPY 2013:485-547. [DOI: 10.1007/978-1-62703-417-3_21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|