1
|
Abdallah AN, Shamaa AA, El-Tookhy OS, Bahr MM. Comparison between stem cell therapy and stem cell derived exosomes on induced multiple sclerosis in dogs. BMC Vet Res 2024; 20:90. [PMID: 38459498 PMCID: PMC10921795 DOI: 10.1186/s12917-024-03920-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 02/08/2024] [Indexed: 03/10/2024] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is a chronic condition that primarily manifests as demyelination of neuronal axons in the central nervous system, due to the loss or attack of oligodendroglia cells that form myelin. Stem cell therapy has shown promising results for the treatment of MS due to its capability to halt the immune attack, stop apoptosis and axonal degeneration, and differentiate into oligodendrocytes. Stem cell-derived Exosomes (Exosomes) have shown great capabilities for neuronal diseases as they have growth factors, complex sets of miRNA, enzymes, proteins, major peptides, lipids, and macromolecules with anti-inflammatory, angiogenesis, and neurogenesis activities. METHODS This study aimed to compare the healing properties of stem cells, against Exosomes for the treatment of an experimentally induced MS dog model. Dog models of MS received either a single treatment of stem cells or a single treatment of Exosomes intrathecally and the treatment process was evaluated clinically, radiologically, histopathologically, and electron microscopy and cerebrospinal fluid analysis. RESULTS showed marked amelioration of the clinical signs in both treated groups compared to the control one, magnetic resonance scans showed the resolution of the hyperintense lesions at the end of the study period, the histopathology and electron microscopy showed marked healing properties and remyelination in treated groups with superiority of the stem cells compared to Exosomes. CONCLUSIONS Although stem cell results were superior to Exosomes therapy; Exosomes have proven to be effective and safe important actors in myelin regeneration, and their use in diseases like MS helps to stimulate remyelination.
Collapse
Affiliation(s)
- Ahmed N Abdallah
- Hormones Department, Medical Research and Clinical Studies Institute, National Research Center, Giza, Egypt
| | - Ashraf A Shamaa
- Surgery, Anesthesiology and Radiology Department, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Omar S El-Tookhy
- Surgery, Anesthesiology and Radiology Department, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Mohamed M Bahr
- Surgery, Anesthesiology and Radiology Department, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt.
| |
Collapse
|
2
|
Roman A, Huntemer-Silveira A, Waldron MA, Khalid Z, Blake J, Parr AM, Low WC. Cell Transplantation for Repair of the Spinal Cord and Prospects for Generating Region-Specific Exogenic Neuronal Cells. Cell Transplant 2024; 33:9636897241241998. [PMID: 38590295 PMCID: PMC11005494 DOI: 10.1177/09636897241241998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 03/05/2024] [Accepted: 03/11/2024] [Indexed: 04/10/2024] Open
Abstract
Spinal cord injury (SCI) is associated with currently irreversible consequences in several functional components of the central nervous system. Despite the severity of injury, there remains no approved treatment to restore function. However, with a growing number of preclinical studies and clinical trials, cell transplantation has gained significant potential as a treatment for SCI. Researchers have identified several cell types as potential candidates for transplantation. To optimize successful functional outcomes after transplantation, one key factor concerns generating neuronal cells with regional and subtype specificity, thus calling on the developmental transcriptome patterning of spinal cord cells. A potential source of spinal cord cells for transplantation is the generation of exogenic neuronal progenitor cells via the emerging technologies of gene editing and blastocyst complementation. This review highlights the use of cell transplantation to treat SCI in the context of relevant developmental gene expression patterns useful for producing regionally specific exogenic spinal cells via in vitro differentiation and blastocyst complementation.
Collapse
Affiliation(s)
- Alex Roman
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Department of Neurosurgery, Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| | - Anne Huntemer-Silveira
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Madison A. Waldron
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Zainab Khalid
- Department of Neurosurgery, Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| | - Jeffrey Blake
- Department of Neurosurgery, Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| | - Ann M. Parr
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Department of Neurosurgery, Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| | - Walter C. Low
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Department of Neurosurgery, Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
3
|
Long-Term Effects of Neural Precursor Cell Transplantation on Secondary Injury Processes and Functional Recovery after Severe Cervical Contusion-Compression Spinal Cord Injury. Int J Mol Sci 2021; 22:ijms222313106. [PMID: 34884911 PMCID: PMC8658203 DOI: 10.3390/ijms222313106] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 11/29/2021] [Accepted: 12/02/2021] [Indexed: 01/21/2023] Open
Abstract
Cervical spinal cord injury (SCI) remains a devastating event without adequate treatment options despite decades of research. In this context, the usefulness of common preclinical SCI models has been criticized. We, therefore, aimed to use a clinically relevant animal model of severe cervical SCI to assess the long-term effects of neural precursor cell (NPC) transplantation on secondary injury processes and functional recovery. To this end, we performed a clip contusion-compression injury at the C6 level in 40 female Wistar rats and a sham surgery in 10 female Wistar rats. NPCs, isolated from the subventricular zone of green fluorescent protein (GFP) expressing transgenic rat embryos, were transplanted ten days after the injury. Functional recovery was assessed weekly, and FluoroGold (FG) retrograde fiber-labeling, as well as manganese-enhanced magnetic resonance imaging (MEMRI), were performed prior to the sacrifice of the animals eight weeks after SCI. After cryosectioning of the spinal cords, immunofluorescence staining was conducted. Results were compared between the treatment groups (NPC, Vehicle, Sham) and statistically analyzed (p < 0.05 was considered significant). Despite the severity of the injury, leading to substantial morbidity and mortality during the experiment, long-term survival of the engrafted NPCs with a predominant differentiation into oligodendrocytes could be observed after eight weeks. While myelination of the injured spinal cord was not significantly improved, NPC treated animals showed a significant increase of intact perilesional motor neurons and preserved spinal tracts compared to untreated Vehicle animals. These findings were associated with enhanced preservation of intact spinal cord tissue. However, reactive astrogliosis and inflammation where not significantly reduced by the NPC-treatment. While differences in the Basso–Beattie–Bresnahan (BBB) score and the Gridwalk test remained insignificant, animals in the NPC group performed significantly better in the more objective CatWalk XT gait analysis, suggesting some beneficial effects of the engrafted NPCs on the functional recovery after severe cervical SCI.
Collapse
|
4
|
Halim A, Qu KY, Zhang XF, Huang NP. Recent Advances in the Application of Two-Dimensional Nanomaterials for Neural Tissue Engineering and Regeneration. ACS Biomater Sci Eng 2021; 7:3503-3529. [PMID: 34291638 DOI: 10.1021/acsbiomaterials.1c00490] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The complexity of the nervous system structure and function, and its slow regeneration rate, makes it more difficult to treat compared to other tissues in the human body when an injury occurs. Moreover, the current therapeutic approaches including the use of autografts, allografts, and pharmacological agents have several drawbacks and can not fully restore nervous system injuries. Recently, nanotechnology and tissue engineering approaches have attracted many researchers to guide tissue regeneration in an effective manner. Owing to their remarkable physicochemical and biological properties, two-dimensional (2D) nanomaterials have been extensively studied in the tissue engineering and regenerative medicine field. The great conductivity of these materials makes them a promising candidate for the development of novel scaffolds for neural tissue engineering application. Moreover, the high loading capacity of 2D nanomaterials also has attracted many researchers to utilize them as a drug/gene delivery method to treat various devastating nervous system disorders. This review will first introduce the fundamental physicochemical properties of 2D nanomaterials used in biomedicine and the supporting biological properties of 2D nanomaterials for inducing neuroregeneration, including their biocompatibility on neural cells, the ability to promote the neural differentiation of stem cells, and their immunomodulatory properties which are beneficial for alleviating chronic inflammation at the site of the nervous system injury. It also discusses various types of 2D nanomaterials-based scaffolds for neural tissue engineering applications. Then, the latest progress on the use of 2D nanomaterials for nervous system disorder treatment is summarized. Finally, a discussion of the challenges and prospects of 2D nanomaterials-based applications in neural tissue engineering is provided.
Collapse
Affiliation(s)
- Alexander Halim
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P.R. China
| | - Kai-Yun Qu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P.R. China
| | - Xiao-Feng Zhang
- Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, P.R. China
| | - Ning-Ping Huang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P.R. China
| |
Collapse
|
5
|
Three Growth Factors Induce Proliferation and Differentiation of Neural Precursor Cells In Vitro and Support Cell-Transplantation after Spinal Cord Injury In Vivo. Stem Cells Int 2020; 2020:5674921. [PMID: 32774390 PMCID: PMC7399764 DOI: 10.1155/2020/5674921] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 03/10/2020] [Accepted: 03/16/2020] [Indexed: 11/25/2022] Open
Abstract
Stem cell therapy with neural precursor cells (NPCs) has the potential to improve neuroregeneration after spinal cord injury (SCI). Unfortunately, survival and differentiation of transplanted NPCs in the injured spinal cord remains low. Growth factors have been successfully used to improve NPC transplantation in animal models, but their extensive application is associated with a relevant financial burden and might hinder translation of findings into the clinical practice. In our current study, we assessed the potential of a reduced number of growth factors in different combinations and concentrations to increase proliferation and differentiation of NPCs in vitro. After identifying a “cocktail” (EGF, bFGF, and PDGF-AA) that directed cell fate towards the oligodendroglial and neuronal lineage while reducing astrocytic differentiation, we translated our findings into an in vivo model of cervical clip contusion/compression SCI at the C6 level in immunosuppressed Wistar rats, combining NPC transplantation and intrathecal administration of the growth factors 10 days after injury. Eight weeks after SCI, we could observe surviving NPCs in the injured animals that had mostly differentiated into oligodendrocytes and oligodendrocytic precursors. Moreover, “Stride length” and “Average Speed” in the CatWalk gait analysis were significantly improved 8 weeks after SCI, representing beneficial effects on the functional recovery with NPC transplantation and the administration of the three growth factors. Nevertheless, no effects on the BBB scores could be observed over the course of the experiment and regeneration of descending tracts as well as posttraumatic myelination remained unchanged. However, reactive astrogliosis, as well as posttraumatic inflammation and apoptosis was significantly reduced after NPC transplantation and GF administration. Our data suggest that NPC transplantation is feasible with the use of only EGF, bFGF, and PDGF-AA as supporting growth factors.
Collapse
|
6
|
Zhou Z, Tian X, Mo B, Xu H, Zhang L, Huang L, Yao S, Huang Z, Wang Y, Xie H, Xu L, Zhang H. Adipose mesenchymal stem cell transplantation alleviates spinal cord injury-induced neuroinflammation partly by suppressing the Jagged1/Notch pathway. Stem Cell Res Ther 2020; 11:212. [PMID: 32493480 PMCID: PMC7268310 DOI: 10.1186/s13287-020-01724-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 04/29/2020] [Accepted: 05/11/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The therapeutic effects of adipose-derived mesenchymal stem cell (ADSC) transplantation have been demonstrated in several models of central nervous system (CNS) injury and are thought to involve the modulation of the inflammatory response. However, the exact underlying molecular mechanism is poorly understood. Activation of the Jagged1/Notch signaling pathway is thought to involve inflammatory and gliotic events in the CNS. Here, we elucidated the effect of ADSC transplantation on the inflammatory reaction after spinal cord injury (SCI) and the potential mechanism mediated by Jagged1/Notch signaling pathway suppression. METHODS To evaluate the therapeutic effects of ADSC treatment and the potential inhibitory effects of ADSCs on Notch signaling, mice were subjected to contusion SCI, and GFP-labeled ADSCs were injected into the lesion site immediately after the injury. Locomotor function, spinal cord tissue morphology, and the levels of Notch-related proteins and proinflammatory transcripts were compared between groups. To validate the hypothesis that the therapeutic effects of ADSCs are partly due to Notch1 signaling inhibition, a Jagged1 small interfering RNA (siRNA) was injected into the spinal cord to knock down Jagged1/Notch signaling. Neuronal staining and analyses of microglia/macrophage activation and signaling pathways were performed. RESULTS We demonstrated that ADSCs survived in the injured spinal cord for at least 28 days without differentiating into glial or neuronal elements. ADSC treatment resulted in significant downregulation of proinflammatory mediator expression and reduced ionized calcium-binding adapter molecule 1 (IBA1) and ED-1 staining in the injured spinal cord, eventually improving functional recovery. The augmentation of the Jagged1/Notch signaling pathway after SCI was suppressed by ADSC transplantation. The inhibition of the Jagged1/Notch signaling pathway by Jagged1 siRNA resulted in decreases in SCI-induced proinflammatory cytokines and the activation of microglia and an increase in the survival of neurons. Furthermore, Jagged1 knockdown suppressed the phosphorylation of JAK/STAT3 in astrocytes following SCI. CONCLUSION The results of this study demonstrated that the therapeutic effects of ADSCs in SCI mice were partly due to Jagged1/Notch signaling pathway inhibition and a subsequent reduction in JAK/STAT3 phosphorylation in astrocytes.
Collapse
Affiliation(s)
- Zhilai Zhou
- The Spine Department, Orthopaedic Center, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Xiaobo Tian
- The Spine Department, Orthopaedic Center, Guangdong Second Provincial General Hospital, Guangzhou, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Biling Mo
- Department of Cardiology, Liwan Central Hospital of Gaungzhou, Guangzhou, China
| | - Huali Xu
- Department of Anesthesiology, Zhu Jiang Hospital, Southern Medical University, Guangzhou, China
| | - Li Zhang
- The Spine Department, Orthopaedic Center, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Lishan Huang
- The Spine Department, Orthopaedic Center, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Shun Yao
- The Spine Department, Orthopaedic Center, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Zixiang Huang
- The Spine Department, Orthopaedic Center, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Yeyang Wang
- The Spine Department, Orthopaedic Center, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Huan Xie
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Liwei Xu
- The Spine Department, Orthopaedic Center, Guangdong Second Provincial General Hospital, Guangzhou, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Hui Zhang
- The Spine Department, Orthopaedic Center, Guangdong Second Provincial General Hospital, Guangzhou, China. .,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.
| |
Collapse
|
7
|
Mahmoodi N, Ai J, Ebrahimi‐Barough S, Hassannejad Z, Hasanzadeh E, Basiri A, Vaccaro AR, Rahimi‐Movaghar V. Microtubule stabilizer epothilone B as a motor neuron differentiation agent for human endometrial stem cells. Cell Biol Int 2020; 44:1168-1183. [DOI: 10.1002/cbin.11315] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 02/02/2020] [Indexed: 12/13/2022]
Affiliation(s)
- Narges Mahmoodi
- Sina Trauma and Surgery Research Center, Sina HospitalTehran University of Medical Sciences Hasan‐Abad Square, Imam Khomeini Ave. Tehran 11365‐3876 Iran
| | - Jafar Ai
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in MedicineTehran University of Medical Sciences Number 88, Italy Street, Between Ghods Street and Vesal Shirazi Street Tehran 14177‐55469 Iran
| | - Somayeh Ebrahimi‐Barough
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in MedicineTehran University of Medical Sciences Number 88, Italy Street, Between Ghods Street and Vesal Shirazi Street Tehran 14177‐55469 Iran
| | - Zahra Hassannejad
- Pediatric Urology and Regenerative Medicine Research Center, Children's Medical Center, Pediatric Center of ExcellenceTehran University of Medical Sciences No. 62, Dr. Gharibs Street, Keshavarz Boulevard Tehran 1419733151 Iran
| | - Elham Hasanzadeh
- Department of Tissue Engineering, School of Advanced Technologies in MedicineMazandaran University of Medical Sciences Next to Tooba Medical Building, Khazar Boulevard Sari 48471‐91971 Iran
| | - Arefeh Basiri
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in MedicineTehran University of Medical Sciences Number 88, Italy Street, Between Ghods Street and Vesal Shirazi Street Tehran 14177‐55469 Iran
| | - Alexander R. Vaccaro
- Department of Orthopedic Surgery, Rothman InstituteThomas Jefferson University 1925 Chestnut Street, 5th Floor Philadelphia Pennsylvania 19107 USA
| | - Vafa Rahimi‐Movaghar
- Sina Trauma and Surgery Research Center, Sina HospitalTehran University of Medical Sciences Hasan‐Abad Square, Imam Khomeini Ave. Tehran 11365‐3876 Iran
| |
Collapse
|
8
|
Hu J, Wang J. From embryonic stem cells to induced pluripotent stem cells-Ready for clinical therapy? Clin Transplant 2019; 33:e13573. [PMID: 31013374 DOI: 10.1111/ctr.13573] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 04/18/2019] [Indexed: 01/08/2023]
Abstract
Embryonic stem cells and induced pluripotent stem cells have increasingly important roles in many different fields of research and medicine. Major areas of impact include improved in vitro disease models, drug screening, and the development of cell-based clinical therapies. Here, we review the generation and uses of embryonic stem cells compared to induced pluripotent stem cells and discuss their advantages and limitations. We also evaluate the feasibility of clinical therapies and the future prospects for induced pluripotent cell-based treatments.
Collapse
Affiliation(s)
- Jing Hu
- Department of Neonatology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Jimei Wang
- Department of Neonatology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| |
Collapse
|
9
|
Fu H, Hu D, Zhang L, Shen X, Tang P. Efficacy of Oligodendrocyte Progenitor Cell Transplantation in Rat Models with Traumatic Thoracic Spinal Cord Injury: A Systematic Review and Meta-Analysis. J Neurotrauma 2018; 35:2507-2518. [PMID: 29759026 DOI: 10.1089/neu.2017.5606] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022] Open
Affiliation(s)
- Haitao Fu
- School of Medicine, Nankai University, Tianjin, China
- Department of Orthopedics, the General Hospital of Chinese People's Liberation Army, Beijing, China
| | - Die Hu
- Qingdao Eye Hospital, Shandong Eye Institute, Shandong Academy of Medical Sciences, Qingdao, China
| | - Licheng Zhang
- Department of Orthopedics, the General Hospital of Chinese People's Liberation Army, Beijing, China
| | - Xuezhen Shen
- School of Medicine, Nankai University, Tianjin, China
- Department of Orthopedics, the General Hospital of Chinese People's Liberation Army, Beijing, China
| | - Peifu Tang
- School of Medicine, Nankai University, Tianjin, China
- Department of Orthopedics, the General Hospital of Chinese People's Liberation Army, Beijing, China
| |
Collapse
|
10
|
Abstract
The history of stem cell therapies is one of a limited number of clinical applications despite a vast therapeutic potential. Major breakthroughs in stem cell research have not yet enjoyed clinical success—all stem cell therapies bar hematopoietic stem cell transplantations remain experimental. With the increased risk of organ failure and neurodegenerative disease associated with our ability to push the boundaries of life expectancy comes an increased pressure to pioneer novel stem cell-based therapeutic approaches. We conclude that the failure of such therapies to achieve clinical translation stems from the polarising effect of the ethical debate around their use. The intractability of the ethical debate is double edged: legislators not only have placed tighter restrictions on certain stem cell therapies, but do so in favour of less controversial cells which will have worse outcomes for patients. It is by considering this relationship between the politics, ethics and science of stem cells that the reasons for the currently limited clinical significance of stem cell therapies be realised.
Collapse
Affiliation(s)
- Jordan Poulos
- University College London Medical School, London, UK.
| |
Collapse
|
11
|
Libro R, Bramanti P, Mazzon E. The combined strategy of mesenchymal stem cells and tissue-engineered scaffolds for spinal cord injury regeneration. Exp Ther Med 2017; 14:3355-3368. [PMID: 29042919 PMCID: PMC5639409 DOI: 10.3892/etm.2017.4939] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 08/03/2017] [Indexed: 01/02/2023] Open
Abstract
Spinal cord injury (SCI) is a traumatic lesion that can result in the loss of motor or sensory neurons. Stem cell (SC)-based therapies have been demonstrated to promote neuronal regeneration following SCI, by releasing a range of trophic factors that support endogenous repair or by differentiating into neurons, or glial cells in order to replace the damaged cells. However, numerous limitations remain for therapies based on SC transplantion alone, including a low rate of survival/engraftment. Nevertheless, scaffolds are 3-dimentional substrates that have revealed to support cell survival, proliferation and differentiation in vivo, by mimicking a more favorable endogenous microenvironment. A multidisciplinary approach, which combines engineered scaffolds with SCs has been proposed as a promising strategy for encouraging spinal cord regeneration. The present review has focused on the regenerative potential of mesenchymal SCs isolated from different sources and combined with various scaffold types, in preclinical and clinical SCI studies.
Collapse
Affiliation(s)
- Rosaliana Libro
- Department of Experimental Neurology, IRCCS Centro Neurolesi ‘Bonino-Pulejo’, I-98124 Messina, Italy
| | - Placido Bramanti
- Department of Experimental Neurology, IRCCS Centro Neurolesi ‘Bonino-Pulejo’, I-98124 Messina, Italy
| | - Emanuela Mazzon
- Department of Experimental Neurology, IRCCS Centro Neurolesi ‘Bonino-Pulejo’, I-98124 Messina, Italy
| |
Collapse
|
12
|
Melo FR, Bressan RB, Forner S, Martini AC, Rode M, Delben PB, Rae GA, Figueiredo CP, Trentin AG. Transplantation of Human Skin-Derived Mesenchymal Stromal Cells Improves Locomotor Recovery After Spinal Cord Injury in Rats. Cell Mol Neurobiol 2017; 37:941-947. [PMID: 27510317 DOI: 10.1007/s10571-016-0414-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 08/02/2016] [Indexed: 12/11/2022]
Abstract
Spinal cord injury (SCI) is a devastating neurologic disorder with significant impacts on quality of life, life expectancy, and economic burden. Although there are no fully restorative treatments yet available, several animal and small-scale clinical studies have highlighted the therapeutic potential of cellular interventions for SCI. Mesenchymal stem cells (MSCs)-which are conventionally isolated from the bone marrow-recently emerged as promising candidates for treating SCI and have been shown to provide trophic support, ameliorate inflammatory responses, and reduce cell death following the mechanical trauma. Here we evaluated the human skin as an alternative source of adult MSCs suitable for autologous cell transplantation strategies for SCI. We showed that human skin-derived MSCs (hSD-MSCs) express a range of neural markers under standard culture conditions and are able to survive and respond to neurogenic stimulation in vitro. In addition, using histological analysis and behavioral assessment, we demonstrated as a proof-of-principle that hSD-MSC transplantation reduces the severity of tissue loss and facilitates locomotor recovery in a rat model of SCI. Altogether, the study provides further characterization of skin-derived MSC cultures and indicates that the human skin may represent an attractive source for cell-based therapies for SCI and other neurological disorders. Further investigation is needed to elucidate the mechanisms by which hSD-MSCs elicit tissue repair and/or locomotor recovery.
Collapse
Affiliation(s)
- Fernanda Rosene Melo
- Departamento de Biologia Celular, Embriologia e Genética, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Campus Universitário, Trindade, Florianópolis, SC, 88040-900, Brazil
| | - Raul Bardini Bressan
- MRC Centre for Regenerative Medicine, The University of Edinburgh, Edinburgh, UK
| | - Stefânia Forner
- Departamento de Farmacologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Campus Universitário, Trindade, Florianópolis, SC, Brazil
| | - Alessandra Cadete Martini
- Departamento de Farmacologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Campus Universitário, Trindade, Florianópolis, SC, Brazil
| | - Michele Rode
- Departamento de Biologia Celular, Embriologia e Genética, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Campus Universitário, Trindade, Florianópolis, SC, 88040-900, Brazil
| | - Priscilla Barros Delben
- Departamento de Biologia Celular, Embriologia e Genética, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Campus Universitário, Trindade, Florianópolis, SC, 88040-900, Brazil
| | - Giles Alexander Rae
- Departamento de Farmacologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Campus Universitário, Trindade, Florianópolis, SC, Brazil
| | - Claudia Pinto Figueiredo
- Faculdade de Farmácia, Centro de Ciências Da Saúde, Universidade Federal Do Rio de Janeiro, Campus Universitário, Ilha do Fundão, Rio de Janeiro, RJ, Brazil
| | - Andrea Gonçalves Trentin
- Departamento de Biologia Celular, Embriologia e Genética, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Campus Universitário, Trindade, Florianópolis, SC, 88040-900, Brazil.
| |
Collapse
|
13
|
Razavi S, Ghasemi N, Mardani M, Salehi H. Remyelination improvement after neurotrophic factors secreting cells transplantation in rat spinal cord injury. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2017; 20:392-398. [PMID: 28804608 PMCID: PMC5425921 DOI: 10.22038/ijbms.2017.8580] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Accepted: 01/12/2017] [Indexed: 01/26/2023]
Abstract
OBJECTIVES Neurotrophic factors secreting cells (NTS-SCs) may be a superior cell source for cell-based therapy in neurodegenerative diseases. NTS-SCs are able to secrete some neurotrophic Such as nerve growth factor and glia-derived neurotrophic factor. Our primary aim was to assess transplantation of neurotrophic factor secreting cells derived from human adipose-derived stem cells (hADSCs) into the damaged spinal cord rats and determine the potential of these cells in remyelination. MATERIALS AND METHODS To this end, 40 adult male Wistar rats were categorized into four groups including; control, lysolecithin (Lysophosphatidylcholines or LPC), vehicle, and NTS-SCs transplan-tation. Local demyelination was induced using LPC injection into the lateral column of spinal cord. Seven days after the lysolecithin lesion, the cells transplantation was performed. The ultrastructure of myelinated fibers was examined with a transmission electron microscope to determine the extent of myelin destruction and remyelinization 4 weeks post cell transplantation. Moreover, the presence of oligodendrocyte in the lesion of spinal cord was assessed by immunohistochemistry procedure. RESULTS The results of current study indicated that in NTF-SCs transplantation group, the remyelination process and the mean of myelin sheath thickness as well as axonal diameters were significantly higher than other groups (P<0.001). Furthermore, immunohistochemistry analysis revealed that in NTF-SCs transplantation group more than 10 percent of transplanted cells were positive for specific markers of oligodendrocyte cells. CONCLUSION NTF-SCs transplantation represents a valuable option for cell-based therapy in the nervous tissue damages.
Collapse
Affiliation(s)
- Shahnaz Razavi
- Department of Anatomical Science, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nazem Ghasemi
- Department of Anatomical Science, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Mardani
- Department of Anatomical Science, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hossein Salehi
- Department of Anatomical Science, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
14
|
Acquisition and Expansion of Adult Rat Bone Marrow Multipotent Mesenchymal Stromal Cells. FOLIA VETERINARIA 2017. [DOI: 10.1515/fv-2017-0003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Abstract
This study was initiated in order to test a mini-invasive method of mesenchymal stem/progenitor cells (MS/PCs) isolation from a rat bone marrow (BM), and subsequently their expansion, differentiation, and evaluation of their immunophenotypic characteristics; and later their preservation as donor cells in an optimal condition for potential autotransplantation. The study group comprised of 6 adult male Sprague-Dawley (S-D) rats, weighing 480—690 g. The rats were anaesthetised by isoflurane with room air in a Plexiglas box and maintained by inhalation of a mixture of isoflurane and O2. Their femurs were surgically exposed and their diaphyses double-trephined. Then BM cells were flushed out by saline with heparin and aspirated into a syringe with a solution of DMEM (Dulbecco’s modified eagle’s medium) and heparin. The mononuclear cells from the BM were isolated by centrifugation and expanded in a standard culture medium supplemented with ES-FBS (es-cell-qualified foetal bovine serum), L-glutamine and rh LIF (recombinant human leukemia inhibitory factor). Following 14 days of passaging cultures, the cells were split into 2 equal parts. The first culture continued with the original medium. The second culture received additional supplementation with a human FGFβ (fibroblast growth factor beta) and EGF (epidermal growth factor). The populations of these cells were analysed by light-microscopy, then the mean fluorescence intensities (MFIs) of CD90 and Nestin were evaluated by a tricolour flow cytometry using monoclonal antibodies. The type of general anaesthesia used proved to be appropriate for the surgical phase of the experiments. All rats survived the harvesting of the BM without complications. The total number of mononuclear cells was 1.5—4.0 × 106 per sample and the proportion of CD90/Nestin expressing cells was < 1 %. Following 14 days of expansion, the cells became larger, adherent, with fibrillary morphology; the proportion of cells expressing CD90/Nestin increased to almost 25 %, i. e. they earned basic phenotypic characteristics of MSCs. Throughout the further cultivation a gradual decrease of the CD90/Nestin expression occurred. This suggested that the suitability of rat bone marrow derived MS/PCs for replacement therapy would probably be the highest between days 12—15 of cultivation and then would diminish.
Collapse
|
15
|
|
16
|
Iyer SR, Xu S, Stains JP, Bennett CH, Lovering RM. Superparamagnetic Iron Oxide Nanoparticles in Musculoskeletal Biology. TISSUE ENGINEERING PART B-REVIEWS 2017; 23:373-385. [PMID: 27998240 DOI: 10.1089/ten.teb.2016.0437] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The use of platelet-rich plasma and mesenchymal stem cells has garnered much attention in orthopedic medicine, focusing on the biological aspects of cell function. However, shortly after systemic delivery, or even a local injection, few of the transplanted stem cells or platelets remain at the target site. Improvement in delivery, and the ability to track and monitor injected cells, would greatly improve clinical translation. Nanoparticles can effectively and quickly label most cells in vitro, and evidence to date suggests such labeling does not compromise the proliferation or differentiation of cells. A specific type of nanoparticle, the superparamagnetic iron oxide nanoparticle (SPION), is already employed as a magnetic resonance imaging (MRI) contrast agent. SPIONs can be coupled with cells or bioactive molecules (antibodies, proteins, drugs, etc.) to form an injectable complex for in vivo use. The biocompatibility, magnetic properties, small size, and custom-made surface coatings also enable SPIONs to be used for delivering and monitoring of small molecules, drugs, and cells, specifically to muscle, bone, or cartilage. Because SPIONs consist of cores made of iron oxides, targeting of SPIONs to a specific muscle, bone, or joint in the body can be enhanced with the help of applied gradient magnetic fields. Moreover, MRI has a high sensitivity to SPIONs and can be used for noninvasive determination of successful delivery and monitoring distribution in vivo. Gaps remain in understanding how the physical and chemical properties of nanomaterials affect biological systems. Nonetheless, SPIONs hold great promise for regenerative medicine, and progress is being made rapidly toward clinical applications in orthopedic medicine.
Collapse
Affiliation(s)
- Shama R Iyer
- 1 Department of Orthopaedics, University of Maryland School of Medicine , Baltimore, Maryland
| | - Su Xu
- 2 Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine , Baltimore, Maryland
| | - Joseph P Stains
- 1 Department of Orthopaedics, University of Maryland School of Medicine , Baltimore, Maryland
| | - Craig H Bennett
- 1 Department of Orthopaedics, University of Maryland School of Medicine , Baltimore, Maryland
| | - Richard M Lovering
- 1 Department of Orthopaedics, University of Maryland School of Medicine , Baltimore, Maryland.,3 Department of Physiology, University of Maryland School of Medicine , Baltimore, Maryland
| |
Collapse
|
17
|
Hur JW, Cho TH, Park DH, Lee JB, Park JY, Chung YG. Intrathecal transplantation of autologous adipose-derived mesenchymal stem cells for treating spinal cord injury: A human trial. J Spinal Cord Med 2016; 39. [PMID: 26208177 PMCID: PMC5137573 DOI: 10.1179/2045772315y.0000000048] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
CONTEXT Spinal cord injury (SCI) can cause irreversible damage to neural tissues. However, there is currently no effective treatment for SCI. The therapeutic potential of adipose-derived mesenchymal stem cells (ADMSCs) has been emerged. OBJECTIVE We evaluated the effects and safety of the intrathecal transplantation of autologous ADMSCs in patients with SCI. Participants/Interventions: Fourteen patients with SCI were enrolled (12 for ASIA A, 1 for B, and 1 for D; duration of impairments 3-28 months). Six patients were injured at cervical, 1 at cervico-thoracic, 6 at thoracic, and 1 at lumbar level. Autologous ADMSCs were isolated from lipoaspirates of patients' subcutaneous fat tissue and 9 × 107 ADMSCs per patient were administered intrathecally through lumbar tapping. MRI, hematological parameters, electrophysiology studies, and ASIA motor/sensory scores were assessed before and after transplantation. RESULTS ASIA motor scores were improved in 5 patients at 8 months follow-up (1-2 grades at some myotomes). Voluntary anal contraction improvement was seen in 2 patients. ASIA sensory score recovery was seen in 10, although degeneration was seen in 1. In somatosensory evoked potential test, one patient showed median nerve improvement. There was no interval change of MRI between baseline and 8 months post-transplantation. Four adverse events were observed in three patients: urinary tract infection, headache, nausea, and vomiting. CONCLUSIONS Over the 8 months of follow-up, intrathecal transplantation of autologous ADMSCs for SCI was free of serious adverse events, and several patients showed mild improvements in neurological function. Patient selection, dosage, and delivery method of ADMSCs should be investigated further.
Collapse
Affiliation(s)
| | - Tai-Hyoung Cho
- Correspondence to: Tai-Hyoung Cho, Department of Neurosurgery, Korea University College of Medicine, Korea University Anam Hospital, #126, 5-Ga, Anam-Dong, Sungbuk-Ku, Seoul, 136-705, Korea.
| | | | | | | | | |
Collapse
|
18
|
Using Stem Cells to Grow Artificial Tissue for Peripheral Nerve Repair. Stem Cells Int 2016; 2016:7502178. [PMID: 27212954 PMCID: PMC4861803 DOI: 10.1155/2016/7502178] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 02/17/2016] [Accepted: 03/02/2016] [Indexed: 12/17/2022] Open
Abstract
Peripheral nerve injury continues to pose a clinical hurdle despite its frequency and advances in treatment. Unlike the central nervous system, neurons of the peripheral nervous system have a greater ability to regenerate. However, due to a number of confounding factors, this is often both incomplete and inadequate. The lack of supportive Schwann cells or their inability to maintain a regenerative phenotype is a major factor. Advances in nervous system tissue engineering technology have led to efforts to build Schwann cell scaffolds to overcome this and enhance the regenerative capacity of neurons following injury. Stem cells that can differentiate along a neural lineage represent an essential resource and starting material for this process. In this review, we discuss the different stem cell types that are showing promise for nervous system tissue engineering in the context of peripheral nerve injury. We also discuss some of the biological, practical, ethical, and commercial considerations in using these different stem cells for future clinical application.
Collapse
|
19
|
Therapeutic potential of human olfactory bulb neural stem cells for spinal cord injury in rats. Spinal Cord 2016; 54:785-797. [PMID: 26882489 DOI: 10.1038/sc.2016.14] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 11/27/2015] [Accepted: 01/05/2016] [Indexed: 12/12/2022]
Abstract
STUDY DESIGN Adult human olfactory bulb neural stem cells (OBNSCs) were isolated from human patients undergoing craniotomy for tumor resection. They were genetically engineered to overexpresses green fluorescent protein (GFP) to help trace them following engraftment. Spinal cord injury (SCI) was induced in rats using standard laminectomy protocol, and GFP-OBNSC were engrafted into rat model of SCI at day 7 post injury. Three rat groups were used: (i) Control group, (ii) Sham group (injected with cerebrospinal fluid) and treated group (engrafted with OBNSCs). Tissues from different groups were collected weekly up to 2 months. The collected tissues were fixed in 4% paraformaldehyde, processed for paraffin sectioning, immunohistochemically stained for different neuronal and glial markers and examined with bright-field fluorescent microscopy. Restoration of sensory motor functions we assessed on a weekly bases using the BBB score. OBJECTIVES To assess the therapeutic potential of OBNSCs-GFP and their ability to survive, proliferate, differentiate and to restore lost sensory motor functions following their engraftment in spinal cord injury (SCI). METHODS GFP-OBNSC were engrafted into a rat model of SCI at day 7 post injury and were followed-up to 8 weeks using behavioral and histochemical methods. RESULTS All transplanted animals exhibited successful engraftment. The survival rate was about 30% of initially transplanted cells. Twenty-seven percent of the engrafted cells differentiated along the NG2 and O4-positive oligodendrocyte lineage, 16% into MAP2 and β-tubulin-positive neurons, and 56% into GFAP-positive astrocytes. CONCLUSION GFP-OBNSCs had survived for >8 weeks after engraftment and were differentiated into neurons, astrocytes and oligodendrocytes, The engrafted cells were distributed throughout gray and white matter of the cord with no evidence of abnormal morphology or any mass formation indicative of tumorigenesis. However, the engrafted cells failed to restore lost sensory and motor functions as evident from behavioral analysis using the BBB score test.
Collapse
|
20
|
Karumbaiah L, Enam SF, Brown AC, Saxena T, Betancur MI, Barker TH, Bellamkonda RV. Chondroitin Sulfate Glycosaminoglycan Hydrogels Create Endogenous Niches for Neural Stem Cells. Bioconjug Chem 2015; 26:2336-49. [PMID: 26440046 DOI: 10.1021/acs.bioconjchem.5b00397] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Neural stem cells (NSCs) possess great potential for neural tissue repair after traumatic injuries to the central nervous system (CNS). However, poor survival and self-renewal of NSCs after injury severely limits its therapeutic potential. Sulfated chondroitin sulfate glycosaminoglycans (CS-GAGs) linked to CS proteoglycans (CSPGs) in the brain extracellular matrix (ECM) have the ability to bind and potentiate trophic factor efficacy, and promote NSC self-renewal in vivo. In this study, we investigated the potential of CS-GAG hydrogels composed of monosulfated CS-4 (CS-A), CS-6 (CS-C), and disulfated CS-4,6 (CS-E) CS-GAGs as NSC carriers, and their ability to create endogenous niches by enriching specific trophic factors to support NSC self-renewal. We demonstrate that CS-GAG hydrogel scaffolds showed minimal swelling and degradation over a period of 15 days in vitro, absorbing only 6.5 ± 0.019% of their initial weight, and showing no significant loss of mass during this period. Trophic factors FGF-2, BDNF, and IL10 bound with high affinity to CS-GAGs, and were significantly (p < 0.05) enriched in CS-GAG hydrogels when compared to unsulfated hyaluronic acid (HA) hydrogels. Dissociated rat subventricular zone (SVZ) NSCs when encapsulated in CS-GAG hydrogels demonstrated ∼88.5 ± 6.1% cell viability in vitro. Finally, rat neurospheres in CS-GAG hydrogels conditioned with the mitogen FGF-2 demonstrated significantly (p < 0.05) higher self-renewal when compared to neurospheres cultured in unconditioned hydrogels. Taken together, these findings demonstrate the ability of CS-GAG based hydrogels to regulate NSC self-renewal, and facilitate growth factor enrichment locally.
Collapse
Affiliation(s)
- Lohitash Karumbaiah
- Regenerative Bioscience Center, ADS Complex, The University of Georgia , 425 River Road, Athens, Georgia 30602, United States
| | | | - Ashley C Brown
- Joint Department of Biomedical Engineering NC State University/UNC-Chapel Hill , 4204 B Engineering Building III, Raleigh, North Carolina 27695, United States
| | | | - Martha I Betancur
- Regenerative Bioscience Center, ADS Complex, The University of Georgia , 425 River Road, Athens, Georgia 30602, United States
| | | | | |
Collapse
|
21
|
The effect of fetal rat brain extract on morphology of bone marrow-derived mesenchymal stem cells. ACTA ACUST UNITED AC 2015. [DOI: 10.1007/s00580-015-2188-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
22
|
Directing cell therapy to anatomic target sites in vivo with magnetic resonance targeting. Nat Commun 2015; 6:8009. [PMID: 26284300 PMCID: PMC4568295 DOI: 10.1038/ncomms9009] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Accepted: 07/08/2015] [Indexed: 01/17/2023] Open
Abstract
Cell-based therapy exploits modified human cells to treat diseases but its targeted application in specific tissues, particularly those lying deep in the body where direct injection is not possible, has been problematic. Here we use a magnetic resonance imaging (MRI) system to direct macrophages carrying an oncolytic virus, Seprehvir, into primary and metastatic tumour sites in mice. To achieve this, we magnetically label macrophages with super-paramagnetic iron oxide nanoparticles and apply pulsed magnetic field gradients in the direction of the tumour sites. Magnetic resonance targeting guides macrophages from the bloodstream into tumours, resulting in increased tumour macrophage infiltration and reduction in tumour burden and metastasis. Our study indicates that clinical MRI scanners can not only track the location of magnetically labelled cells but also have the potential to steer them into one or more target tissues. Cell therapy requires the targeting of cells to specific sites in the body. Here Muthana et al. use a standard MRI scanner to direct oncolytic macrophages, labelled with magnetic nanoparticles, to primary and metastatic tumour sites in mice, and demonstrate that this leads to reduced tumour growth.
Collapse
|
23
|
Rak K, Völker J, Jürgens L, Völker C, Frenz S, Scherzad A, Schendzielorz P, Jablonka S, Mlynski R, Radeloff A, Hagen R. Cochlear nucleus whole mount explants promote the differentiation of neuronal stem cells from the cochlear nucleus in co-culture experiments. Brain Res 2015; 1616:58-70. [PMID: 25960344 DOI: 10.1016/j.brainres.2015.04.055] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 04/26/2015] [Accepted: 04/29/2015] [Indexed: 10/23/2022]
Abstract
The cochlear nucleus is the first brainstem nucleus to receive sensory input from the cochlea. Depriving this nucleus of auditory input leads to cellular and molecular disorganization which may potentially be counteracted by the activation or application of stem cells. Neuronal stem cells (NSCs) have recently been identified in the neonatal cochlear nucleus and a persistent neurogenic niche was demonstrated in this brainstem nucleus until adulthood. The present work investigates whether the neurogenic environment of the cochlear nucleus can promote the survival of engrafted NSCs and whether cochlear nucleus-derived NSCs can differentiate into neurons and glia in brain tissue. Therefore, cochlear nucleus whole-mount explants were co-cultured with NSCs extracted from either the cochlear nucleus or the hippocampus and compared to a second environment using whole-mount explants from the hippocampus. Factors that are known to induce neuronal differentiation were also investigated in these NSC-explant experiments. NSCs derived from the cochlear nucleus engrafted in the brain tissue and differentiated into all cells of the neuronal lineage. Hippocampal NSCs also immigrated in cochlear nucleus explants and differentiated into neurons, astrocytes and oligodendrocytes. Laminin expression was up-regulated in the cochlear nucleus whole-mounts and regulated the in vitro differentiation of NSCs from the cochlear nucleus. These experiments confirm a neurogenic environment in the cochlear nucleus and the capacity of cochlear nucleus-derived NSCs to differentiate into neurons and glia. Consequently, the presented results provide a first step for the possible application of stem cells to repair the disorganization of the cochlear nucleus, which occurs after hearing loss.
Collapse
Affiliation(s)
- Kristen Rak
- Department of Oto-Rhino-Laryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery and the Comprehensive Hearing Center, University of Wuerzburg, Wuerzburg, Germany.
| | - Johannes Völker
- Department of Oto-Rhino-Laryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery and the Comprehensive Hearing Center, University of Wuerzburg, Wuerzburg, Germany
| | - Lukas Jürgens
- Department of Oto-Rhino-Laryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery and the Comprehensive Hearing Center, University of Wuerzburg, Wuerzburg, Germany
| | - Christine Völker
- Department of Oto-Rhino-Laryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery and the Comprehensive Hearing Center, University of Wuerzburg, Wuerzburg, Germany
| | - Silke Frenz
- Department of Oto-Rhino-Laryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery and the Comprehensive Hearing Center, University of Wuerzburg, Wuerzburg, Germany
| | - Agmal Scherzad
- Department of Oto-Rhino-Laryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery and the Comprehensive Hearing Center, University of Wuerzburg, Wuerzburg, Germany
| | - Philipp Schendzielorz
- Department of Oto-Rhino-Laryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery and the Comprehensive Hearing Center, University of Wuerzburg, Wuerzburg, Germany
| | - Sibylle Jablonka
- Institute for Clinical Neurobiology, University of Wuerzburg, Wuerzburg, Germany
| | - Robert Mlynski
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery "Otto Körner", Rostock University Medical Center, Rostock, Germany
| | - Andreas Radeloff
- Department of Oto-Rhino-Laryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery and the Comprehensive Hearing Center, University of Wuerzburg, Wuerzburg, Germany
| | - Rudolf Hagen
- Department of Oto-Rhino-Laryngology, Plastic, Aesthetic and Reconstructive Head and Neck Surgery and the Comprehensive Hearing Center, University of Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
24
|
Wang Z, Xia J, Yan Y, Tsai AC, Li Y, Ma T, Guan J. Facile functionalization and assembly of live cells with microcontact-printed polymeric biomaterials. Acta Biomater 2015; 11:80-7. [PMID: 25305514 DOI: 10.1016/j.actbio.2014.10.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 09/23/2014] [Accepted: 10/02/2014] [Indexed: 01/11/2023]
Abstract
The functionalization and assembly of live cells with microfabricated polymeric biomaterials have attracted considerable interest in recent years, but the conventional methods suffer from high cost, high complexity, long processing time or inadequate capability. The present study reports on the development of a novel method for functionalizing and assembling live cells by integrating microcontact printing of polymeric biomaterials with a temperature-sensitive sacrificial layer prepared by spin-coating. This method has been used not only to functionalize live cells with microscopic polyelectrolyte and thermoplastic structures of various sizes and shapes, but also to assemble the cells into macroscopic stripes and sheets. The method is applicable to multiple types of cells, including human leukemic cells, mouse embryonic stem cells and human mesenchymal stem cells in the forms of single cells and cell aggregates. In addition, the microcontact-printed structures can be prepared using biodegradable and biocompatible polyelectrolytes and thermoplastic. The unique combination of low cost, ease of use and high versatility renders this method potentially useful for diverse biomedical applications, including drug delivery, cell tracking and tissue engineering.
Collapse
|
25
|
Survival of neural stem cell grafts in the lesioned spinal cord is enhanced by a combination of treadmill locomotor training via insulin-like growth factor-1 signaling. J Neurosci 2014; 34:12788-800. [PMID: 25232115 DOI: 10.1523/jneurosci.5359-13.2014] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Combining cell transplantation with activity-based rehabilitation is a promising therapeutic approach for spinal cord repair. The present study was designed to investigate potential interactions between the transplantation (TP) of neural stem cells (NSCs) obtained at embryonic day 14 and treadmill training (TMT) in promoting locomotor recovery and structural repair in rat contusive injury model. Combination of TMT with NSC TP at 1 week after injury synergistically improved locomotor function. We report here that combining TMT increased the survival of grafted NSCs by >3-fold and >5-fold at 3 and 9 weeks after injury, respectively. The number of surviving NSCs was significantly correlated with the extent of locomotor recovery. NSCs grafted into the injured spinal cord were under cellular stresses induced by reactive nitrogen or oxygen species, which were markedly attenuated by TMT. TMT increased the concentration of insulin-like growth factor-1 (IGF-1) in the CSF. Intrathecal infusion of neutralizing IGF-1 antibodies, but not antibodies against either BDNF or Neurotrophin-3 (NT-3), abolished the enhanced survival of NSC grafts by TMT. The combination of TP and TMT also resulted in tissue sparing, increased myelination, and restoration of serotonergic fiber innervation to the lumbar spinal cord to a larger extent than that induced by either TP or TMT alone. Therefore, we have discovered unanticipated beneficial effects of TMT in modulating the survival of grafted NSCs via IGF-1. Our study identifies a novel neurobiological basis for complementing NSC-based spinal cord repair with activity-based neurorehabilitative approaches.
Collapse
|
26
|
Evaluation of Motor Neuron-Like Cell Differentiation of hEnSCs on Biodegradable PLGA Nanofiber Scaffolds. Mol Neurobiol 2014; 52:1704-1713. [DOI: 10.1007/s12035-014-8931-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 10/07/2014] [Indexed: 10/24/2022]
|
27
|
Cherry JF, Bennett NK, Schachner M, Moghe PV. Engineered N-cadherin and L1 biomimetic substrates concertedly promote neuronal differentiation, neurite extension and neuroprotection of human neural stem cells. Acta Biomater 2014; 10:4113-26. [PMID: 24914828 DOI: 10.1016/j.actbio.2014.06.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 04/26/2014] [Accepted: 06/01/2014] [Indexed: 02/05/2023]
Abstract
We investigated the design of neurotrophic biomaterial constructs for human neural stem cells, guided by neural developmental cues of N-cadherin and L1 adhesion molecules. Polymer substrates fabricated either as two-dimensional (2-D) films or three-dimensional (3-D) microfibrous scaffolds were functionalized with fusion chimeras of N-cadherin-Fc alone and in combination with L1-Fc, and the effects on differentiation, neurite extension and survival of H9 human-embryonic-stem-cell-derived neural stem cells (H9-NSCs) were quantified. Combinations of N-cadherin and L1-Fc co-operatively enhanced neuronal differentiation profiles, indicating the critical nature of the two complementary developmental cues. Notably, substrates presenting low levels of N-cadherin-Fc concentrations, combined with proportionately higher L1-Fc concentration, most enhanced neurite outgrowth and the degree of MAP2+ and neurofilament-M+ H9-NSCs. Low N-cadherin-Fc alone promoted improved cell survival following oxidative stress, compared to higher concentrations of N-cadherin-Fc alone or combinations with L1-Fc. Pharmacological and antibody blockage studies revealed that substrates presenting low levels of N-cadherin are functionally competent so long as they elicit a threshold signal mediated by homophilic N-cadherin and fibroblast growth factor signaling. Overall, these studies highlight the ability of optimal combinations of N-cadherin and L1 to recapitulate a "neurotrophic" microenvironment that enhances human neural stem cell differentiation and neurite outgrowth. Additionally, 3-D fibrous scaffolds presenting low N-cadherin-Fc further enhanced the survival of H9-NSCs compared to equivalent 2-D films. This indicates that similar biofunctionalization approaches based on N-cadherin and L1 can be translated to 3-D "transplantable" scaffolds with enhanced neurotrophic behaviors. Thus, the insights from this study have fundamental and translational impacts for neural-stem-cell-based regenerative medicine.
Collapse
Affiliation(s)
- Jocie F Cherry
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, USA
| | - Neal K Bennett
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, USA
| | - Melitta Schachner
- W.M. Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, USA; Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou 515041, People's Republic of China
| | - Prabhas V Moghe
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, USA; Department of Chemical and Biochemical Engineering, Rutgers University, 599 Taylor Road, Piscataway, NJ 08854, USA.
| |
Collapse
|
28
|
Nishimura Y, Natsume A, Ito M, Hara M, Motomura K, Fukuyama R, Sumiyoshi N, Aoki I, Saga T, Lee HJ, Wakabayashi T, Kim SU. Interferon-β Delivery via Human Neural Stem Cell Abates Glial Scar Formation in Spinal Cord Injury. Cell Transplant 2013; 22:2187-201. [DOI: 10.3727/096368912x657882] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Glial scar formation is the major impedance to axonal regrowth after spinal cord injury (SCI), and scar-modulating treatments have become a leading therapeutic goal for SCI treatment. In this study, human neural stem cells (NSCs) encoding interferon-β (INF-β) gene were administered intravenously to mice 1 week after SCI. Animals receiving NSCs encoding IFN-β exhibited significant neurobehavioral improvement, electrophysiological recovery, suppressed glial scar formation, and preservation of nerve fibers in lesioned spinal cord. Systemic evaluation of SCI gliosis lesion site with lesion-specific microdissection, genome-wide microarray, and MetaCore pathway analysis identified upregulation of toll-like receptor 4 (TLR4) in SCI gliosis lesion site, and this led us to focus on TLR4 signaling in reactive astrocytes. Examination of primary astrocytes from TLR4 knockout mice, and in vivo inhibition of TLR4, revealed that the effect of IFN-β on the suppression of glial scar formation in SCI requires TLR4 stimulation. These results suggest that IFN-β delivery via intravenous injection of NSCs following SCI inhibits glial scar formation in spinal cord through stimulation of TLR4 signaling.
Collapse
Affiliation(s)
| | - Atsushi Natsume
- Department of Neurosurgery, Nagoya University, Nagoya, Japan
| | - Motokazu Ito
- Department of Neurosurgery, Nagoya University, Nagoya, Japan
| | - Masahito Hara
- Department of Neurosurgery, Nagoya University, Nagoya, Japan
| | - Kazuya Motomura
- Department of Neurosurgery, Nagoya University, Nagoya, Japan
| | | | | | - Ichio Aoki
- MR Molecular Imaging Team, Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan
| | - Tsuneo Saga
- MR Molecular Imaging Team, Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan
| | - Hong J. Lee
- Medical Research Institute, Chung-Ang University College of Medicine, Seoul, Korea
| | | | - Seung U. Kim
- Medical Research Institute, Chung-Ang University College of Medicine, Seoul, Korea
- Division of Neurology, Department of Medicine, UBC Hospital, University of British Columbia, Vancouver, Canada
| |
Collapse
|
29
|
Binan L, Tendey C, De Crescenzo G, El Ayoubi R, Ajji A, Jolicoeur M. Differentiation of neuronal stem cells into motor neurons using electrospun poly-L-lactic acid/gelatin scaffold. Biomaterials 2013; 35:664-74. [PMID: 24161168 DOI: 10.1016/j.biomaterials.2013.09.097] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Accepted: 09/24/2013] [Indexed: 12/17/2022]
Abstract
Neural stem cells (NSCs) provide promising therapeutic potential for cell replacement therapy in spinal cord injury (SCI). However, high increases of cell viability and poor control of cell differentiation remain major obstacles. In this study, we have developed a non-woven material made of co-electrospun fibers of poly L-lactic acid and gelatin with a degradation rate and mechanical properties similar to peripheral nerve tissue and investigated their effect on cell survival and differentiation into motor neuronal lineages through the controlled release of retinoic acid (RA) and purmorphamine. Engineered Neural Stem-Like Cells (NSLCs) seeded on these fibers, with and without the instructive cues, differentiated into β-III-tubulin, HB-9, Islet-1, and choactase-positive motor neurons by immunostaining, in response to the release of the biomolecules. In addition, the bioactive material not only enhanced the differentiation into motor neuronal lineages but also promoted neurite outgrowth. This study elucidated that a combination of electrospun fiber scaffolds, neural stem cells, and controlled delivery of instructive cues could lead to the development of a better strategy for peripheral nerve injury repair.
Collapse
Affiliation(s)
- Loïc Binan
- Department of Chemical Engineering, École Polytechnique de Montréal, P.O. Box 6079, Centre-Ville Station, H3C 3A7 Montréal, Québec, Canada; Groupe de Recherche en Science et Technologie Biomédicale (GRSTB), École Polytechnique de Montréal, P.O. Box 6079, Centre-Ville Station, H3C 3A7 Montréal, Québec, Canada
| | | | | | | | | | | |
Collapse
|
30
|
Hurst RW, Bosch EP, Peter Bosch E, Morris JM, Dyck PJB, Reeves RK. Inflammatory hypertrophic cauda equina following intrathecal neural stem cell injection. Muscle Nerve 2013; 48:831-5. [PMID: 23740462 DOI: 10.1002/mus.23920] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/23/2013] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Potential benefit from stem cell treatments has more patients seeking treatment without understanding possible risks. METHODS We describe a woman who presented with progressive bilateral leg pain, numbness, and gait difficulties. A prior stroke, macular degeneration, osteoarthritis, and depression, led her to receive intrathecal neural stem cell therapy overseas 1 year before onset of symptoms. RESULTS Imaging showed marked enlargement of lumbosacral roots of the cauda equina, which was not seen before stem cell treatment. Electrodiagnostic studies confirmed chronic multiple lumbosacral radiculopathies. Biopsy of a lumbar dorsal sensory root showed myelinated fiber degeneration and loss, with endoneurial inflammation. The hypertrophic inflammatory cauda equina syndrome was potentially triggered by the prior intrathecal neural stem cell injection. CONCLUSIONS Safety of intrathecal stem cell treatments is not routinely regulated in overseas stem cell facilities. We wish to bring this potential complication to the attention of health care providers.
Collapse
Affiliation(s)
- Richard W Hurst
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, Minnesota, USA
| | | | | | | | | | | |
Collapse
|
31
|
Mohammad-Gharibani P, Tiraihi T, Delshad A, Arabkheradmand J, Taheri T. Improvement of contusive spinal cord injury in rats by co-transplantation of gamma-aminobutyric acid-ergic cells and bone marrow stromal cells. Cytotherapy 2013; 15:1073-85. [PMID: 23806239 DOI: 10.1016/j.jcyt.2013.05.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Revised: 04/25/2013] [Accepted: 05/07/2013] [Indexed: 01/01/2023]
Abstract
BACKGROUND AIMS Cell therapy is considered a promising option for treatment of spinal cord injury (SCI). The purpose of this study is to use combined therapy of bone marrow stromal cells (BMSCs) and BMSC-derived gamma-aminobutyric acid (GABA)ergic inhibitory neurotransmitter cells (BDGCs) for the contusion model of SCI in rats. METHODS BDGCs were prepared from BMSCs by pre-inducing them with β-mercaptoethanol followed by retinoic acid and then inducing them by creatine. They were immunostained with BMSC, proneuronal, neural and GABA markers. The BDGCs were intraspinally transplanted into the contused rats, whereas the BMSCs were delivered intravenously. The animals were sacrificed after 12 weeks. RESULTS The Basso, Beattie and Bresnahan test showed improvement in the animals with the combined therapy compared with the untreated animals, the animals treated with GABAergic cells only and the animals that received BMSCs. The immunohistochemistry analysis of the tissue sections prepared from the animals receiving the combined therapy showed that the transplanted cells were engrafted and integrated into the injured spinal cord; in addition, a significant reduction was seen in the cavitation. CONCLUSIONS The study shows that the combination of GABAergic cells with BMSCs can improve SCI.
Collapse
Affiliation(s)
- Payam Mohammad-Gharibani
- Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | | | | | | | |
Collapse
|
32
|
Deboux C, Ladraa S, Cazaubon S, Ghribi-Mallah S, Weiss N, Chaverot N, Couraud PO, Evercooren ABV. Overexpression of CD44 in neural precursor cells improves trans-endothelial migration and facilitates their invasion of perivascular tissues in vivo. PLoS One 2013; 8:e57430. [PMID: 23468987 PMCID: PMC3585392 DOI: 10.1371/journal.pone.0057430] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Accepted: 01/22/2013] [Indexed: 02/02/2023] Open
Abstract
Neural precursor (NPC) based therapies are used to restore neurons or oligodendrocytes and/or provide neuroprotection in a large variety of neurological diseases. In multiple sclerosis models, intravenously (i.v) -delivered NPCs reduced clinical signs via immunomodulation. We demonstrated recently that NPCs were able to cross cerebral endothelial cells in vitro and that the multifunctional signalling molecule, CD44 involved in trans-endothelial migration of lymphocytes to sites of inflammation, plays a crucial role in extravasation of syngeneic NPCs. In view of the role of CD44 in NPCs trans-endothelial migration in vitro, we questioned presently the benefit of CD44 overexpression by NPCs in vitro and in vivo, in EAE mice. We show that overexpression of CD44 by NPCs enhanced over 2 folds their trans-endothelial migration in vitro, without impinging on the proliferation or differentiation potential of the transduced cells. Moreover, CD44 overexpression by NPCs improved significantly their elongation, spreading and number of filopodia over the extracellular matrix protein laminin in vitro. We then tested the effect of CD44 overexpression after i.v. delivery in the tail vein of EAE mice. CD44 overexpression was functional invivo as it accelerated trans-endothelial migration and facilitated invasion of HA expressing perivascular sites. These in vitro and in vivo data suggest that CD44 may be crucial not only for NPC crossing the endothelial layer but also for facilitating invasion of extravascular tissues.
Collapse
Affiliation(s)
- Cyrille Deboux
- Université Pierre et Marie Curie-Paris 6, Centre de Recherche de l’Institut du Cerveau et de la Moelle Epinière, UMR-S975, Paris, France
- Inserm, U 975, Paris, France
- CNRS, UMR 7225, Paris, France
| | - Sophia Ladraa
- Université Pierre et Marie Curie-Paris 6, Centre de Recherche de l’Institut du Cerveau et de la Moelle Epinière, UMR-S975, Paris, France
- Inserm, U 975, Paris, France
- CNRS, UMR 7225, Paris, France
| | - Sylvie Cazaubon
- Inserm, U1016, Institut Cochin, Paris, France
- CNRS (UMR8104), Paris Descartes, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Siham Ghribi-Mallah
- Université Pierre et Marie Curie-Paris 6, Centre de Recherche de l’Institut du Cerveau et de la Moelle Epinière, UMR-S975, Paris, France
- Inserm, U 975, Paris, France
- CNRS, UMR 7225, Paris, France
| | - Nicolas Weiss
- Inserm, U1016, Institut Cochin, Paris, France
- CNRS (UMR8104), Paris Descartes, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Nathalie Chaverot
- Inserm, U1016, Institut Cochin, Paris, France
- CNRS (UMR8104), Paris Descartes, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Pierre Olivier Couraud
- Inserm, U1016, Institut Cochin, Paris, France
- CNRS (UMR8104), Paris Descartes, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Anne Baron-Van Evercooren
- Université Pierre et Marie Curie-Paris 6, Centre de Recherche de l’Institut du Cerveau et de la Moelle Epinière, UMR-S975, Paris, France
- Inserm, U 975, Paris, France
- CNRS, UMR 7225, Paris, France
- Assitance Publique-Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Fédération de Neurologie
- * E-mail:
| |
Collapse
|
33
|
Rao Y, Zhu W, Liu H, Jia C, Zhao Q, Wang Y. Clinical application of olfactory ensheathing cells in the treatment of spinal cord injury. J Int Med Res 2013; 41:473-81. [PMID: 23569013 DOI: 10.1177/0300060513476426] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVES To investigate the safety and therapeutic efficacy of autologous olfactory ensheathing cell (OEC) transplantation in cervical spinal cord injury (SCI). METHODS Patients with cervical SCI of >6 months' duration were treated with autologous OECs, injected into the area surrounding the SCI under magnetic resonance imaging guidance, twice a week for 4 weeks. Patients were evaluated before treatment and at 3, 6, 12 and 24 months post-treatment, using the American Spinal Injury Association (ASIA) Impairment Scale, the ASIA sensory and motor score and the Functional Independence Measure (FIM) score. RESULTS Eight patients were recruited to the study. Three months after treatment, ASIA and FIM scores had improved significantly compared with pretreatment, though by 1 year no further significant improvements in the ASIA score were seen. The return of substantial sensation and motor activity in various muscles below the injury level was observed in three patients during follow-up. In addition, bladder function was restored in two patients. There were no serious complications postoperatively or during the follow-up period. CONCLUSIONS This study provides preliminary evidence of the safety and possible efficacy of autologous OEC transplantation.
Collapse
Affiliation(s)
- Yaojian Rao
- Luoyang Orthopaedic-Traumatological Hospital, Luoyang, China
| | | | | | | | | | | |
Collapse
|
34
|
Alternatively Activated Macrophages in Spinal Cord Injury and Remission: Another Mechanism for Repair? Mol Neurobiol 2013; 47:1011-9. [DOI: 10.1007/s12035-013-8398-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Accepted: 01/03/2013] [Indexed: 11/25/2022]
|
35
|
Cao HQ, Dong ED. An update on spinal cord injury research. Neurosci Bull 2012; 29:94-102. [PMID: 23124646 DOI: 10.1007/s12264-012-1277-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Accepted: 07/26/2012] [Indexed: 02/06/2023] Open
Abstract
Spinal cord injury (SCI) can have a range of debilitating effects and permanently alter the capabilities and quality of life of survivors. The first specialized centers of care for SCI were established in 1944 and since then an increasing amount of research has been carried out in this area. Despite this, the present treatment and care levels for SCI are not comparable to those in other areas of medicine. In the clinic, the aim of SCI treatment is primarily to limit secondary damage by reducing compression in trauma spots and stabilizing the spinal column. Currently, no effective strategy for functional recovery is offered. In this review, we focus on research progress on the molecular mechanisms underlying SCI, and assess the treatment outcomes of SCI in animal models, i.e., neurotrophins and stem cells are discussed as pre-clinical therapies in animal models. We also assess the resources available and national research projects carried out on SCI in China in recent years, as well as making recommendations for the future allocation of funds in this area.
Collapse
Affiliation(s)
- He-Qi Cao
- Division of Neurological Disorders and Mental Health, Department of Health Sciences, National Natural Science Foundation of China, Beijing, 100085, China.
| | | |
Collapse
|
36
|
Volarevic V, Erceg S, Bhattacharya SS, Stojkovic P, Horner P, Stojkovic M. Stem cell-based therapy for spinal cord injury. Cell Transplant 2012; 22:1309-23. [PMID: 23043847 DOI: 10.3727/096368912x657260] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Stem cells (SCs) represent a new therapeutic approach for spinal cord injury (SCI) by enabling improved sensory and motor functions in animal models. The main goal of SC-based therapy for SCI is the replacement of neurons and glial cells that undergo cell death soon after injury. Stem cells are able to promote remyelination via oligodendroglia cell replacement to produce trophic factors enhancing neurite outgrowth, axonal elongation, and fiber density and to activate resident or transplanted progenitor cells across the lesion cavity. While several SC transplantation strategies have shown promising yet partial efficacy, mechanistic proof is generally lacking and is arguably the largest impediment toward faster progress and clinical application. The main challenge ahead is to spur on cooperation between clinicians, researchers, and patients in order to define and optimize the mechanisms of SC function and to establish the ideal source/s of SCs that produce efficient and also safe therapeutic approaches.
Collapse
Affiliation(s)
- Vladislav Volarevic
- Center for Molecular Medicine and Stem Cell Research, Medical Faculty, University of Kragujevac, Serbia
| | | | | | | | | | | |
Collapse
|
37
|
Paspala SA, Vishwakarma SK, Murthy TV, Rao TN, Khan AA. Potential role of stem cells in severe spinal cord injury: current perspectives and clinical data. STEM CELLS AND CLONING-ADVANCES AND APPLICATIONS 2012; 5:15-27. [PMID: 24198535 PMCID: PMC3781762 DOI: 10.2147/sccaa.s28477] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Stem cell transplantation for spinal cord injury (SCI) along with new pharmacotherapy research offers the potential to restore function and ease the associated social and economic burden in the years ahead. Various sources of stem cells have been used in the treatment of SCI, but the most convincing results have been obtained with neural progenitor cells in preclinical models. Although the use of cell-based transplantation strategies for the repair of chronic SCI remains the long sought after holy grail, these approaches have been to date the most successful when applied in the subacute phase of injury. Application of cell-based strategies for the repair and regeneration of the chronically injured spinal cord will require a combinational strategy that may need to include approaches to overcome the effects of the glial scar, inhibitory molecules, and use of tissue engineering strategies to bridge the lesion. Nonetheless, cell transplantation strategies are promising, and it is anticipated that the Phase I clinical trials of some form of neural stem cell-based approach in SCI will commence very soon.
Collapse
Affiliation(s)
- Syed Ab Paspala
- PAN Research Foundation, CARE, Hyderabad, India ; The Institute of Medical Sciences, Hyderabad, India
| | | | | | | | | |
Collapse
|
38
|
Xu W, Li P, Qin K, Wang X, Jiang X. miR-124 regulates neural stem cells in the treatment of spinal cord injury. Neurosci Lett 2012; 529:12-7. [PMID: 22999930 DOI: 10.1016/j.neulet.2012.09.025] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Revised: 09/05/2012] [Accepted: 09/12/2012] [Indexed: 01/17/2023]
Abstract
Several studies demonstrated that the overexpression of miR-124 in neural stem cells (NSCs) could lead the NSCs to differentiate into neurons and astrocytes, which may be important for functional recovery in spinal cord injury. The present study attempted to explore the potential repairing effect of the NSCs transfected with miR-124 for the rats with spinal cord injury (SCI). NSCs transfected with miR-124 were transplanted into rats by intravenous injection after SCI. The effects of miR-124 on the differentiation of NSCs and the treatment for the SCI-model rats were experimentally investigated. The reduction of cavity volume in focal lesions and Basso-Beattie-Bresnahan (BBB) scores were used as the criteria of functional recovery of the SCI-model rats. Up-regulation of miR-124 promoted the differentiation of NSCs. Transfection of miR-124 in NSCs dramatically increased the percentage of NeuN-positive cells, and reduced the percentage of GFAP-positive cells in vitro and in vivo respectively. All of the rats treated with NSCs transfected with miR-124 achieved the better functional recovery than the ones in NSCs and sham control groups. Furthermore, the systemic delivery of the NSCs transfected with miR-124 resulted in a reduction of lesion cavity volume of SCI-model rats. Thus, Overexpression of miR-124 can promote the differentiation of NSCs and play an important role in the repair of SCI. The utility of intravenous delivery of stem cells regulated with miR-124 to target lesion areas as a prospective therapeutic approach in acute spinal cord injury is very promising in the future.
Collapse
Affiliation(s)
- Weiwei Xu
- Department of Neurosurgery, The First Affiliated Hospital of Jinan University, Guangdong Province 510630, China
| | | | | | | | | |
Collapse
|
39
|
Zhilai Z, Hui Z, Anmin J, Shaoxiong M, Bo Y, Yinhai C. A combination of taxol infusion and human umbilical cord mesenchymal stem cells transplantation for the treatment of rat spinal cord injury. Brain Res 2012; 1481:79-89. [PMID: 22960115 DOI: 10.1016/j.brainres.2012.08.051] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Revised: 08/14/2012] [Accepted: 08/27/2012] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND PURPOSE Studies have shown that the administration of Taxol, an anti-cancer drug, inhibited scar formation, promoted axonal elongation and improved locomotor recovery in rats after spinal cord injury (SCI). We hypothesized that combining Taxol with another promising therapy, transplantation of human umbilical mesenchymal stem cells (hUCMSCs), might further improve the degree of locomotor recovery. The present study examined whether Taxol combined with transplantation of hUCMSCs would produce synergistic effects on recovery and which mechanisms were involved in the effect. METHODS A total of 32 rats subjected to SCI procedures were assigned to one of the following four treatment groups: phosphate-buffered saline (PBS, control), hUCMSCs, Taxol, or Taxol+hUCMSCs. Immediately after injury, hUCMSCs were transplanted into the injury site and Taxol was administered intrathecally for 4 weeks. Locomotor recovery was evaluated using the Basso, Beattie and Bresnahan locomotor (BBB) rating scale. Survival of the transplanted human cells and the host glial reaction in the injured spinal cord were studied by immunohistochemistry. RESULTS Treatment with Taxol, hUCMSCs or Taxol+hUCMSCs reduced the extent of astrocytic activation, increased axonal preservation and decreased the number of caspase-3(+) and ED-1(+) cells, but these effects were more pronounced in the Taxol+hUCMSCs group. Behavioral analyses showed that rats in the Taxol+hUCMSCs group showed better motor performance than rats treated with hUCMSCs or Taxol only. CONCLUSIONS The combination of Taxol and hUCMSCs produced beneficial effects in rats with regard to functional recovery following SCI through the enhancement of anti-inflammatory, anti-astrogliosis, anti-apoptotic and axonal preservation effects.
Collapse
Affiliation(s)
- Zhou Zhilai
- Department of Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | | | | | | | | | | |
Collapse
|
40
|
|
41
|
Guo X, Zahir T, Mothe A, Shoichet MS, Morshead CM, Katayama Y, Tator CH. The Effect of Growth Factors and Soluble Nogo-66 Receptor Protein on Transplanted Neural Stem/Progenitor Survival and Axonal Regeneration after Complete Transection of Rat Spinal Cord. Cell Transplant 2012; 21:1177-97. [DOI: 10.3727/096368911x612503] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Adult central mammalian axons show minimal regeneration after spinal cord injury due to loss of oligodendrocytes, demyelination of surviving axons, absence of growth-promoting molecules, and inhibitors of axonal outgrowth. In the present study, we attempted to address these impediments to regeneration by using a combinatory strategy to enhance cell survival and regeneration after complete spinal cord transection (SCT) in adult rats. The strategy comprised: 1) adult rat brain-derived neural stem/progenitor cells (NSPCs) preseeded on laminin-coated chitosan channels; 2) extramedullary chitosan channels to promote axonal regrowth and reduce the barrier caused by scarring; 3) local delivery of a novel rat soluble Nogo-66 receptor protein [NgR(310)ecto-Fc, referred to as NgR] to block the inhibitory effect of myelin-based inhibitors; and 4) local delivery of basic fibroblast growth factor, epidermal growth factor, and platelet-derived growth factor to enhance survival and promote differentiation of transplanted cells. Compared with our previous studies where brain-derived NSPCs preseeded in extramedullary chitosan channels were implanted in the same SCT model but without growth factors and NgR, the present channel–growth factor combination produced greater numbers of surviving NSPCs after SCT. Also, the growth factors promoted preferential differentiation of NSPCs toward oligodendrocytes, while NgR significantly decreased astrocytic differentiation of NSPCs. NgR alone or in combination with NSPCs significantly enhanced the total number of myelinated fibers in the bridge and increased the area of the bridging tissue between the cord stumps. The combination of NgR, growth factors, and NSPCs had synergistic effect on bridge formation. However, only a small number of descending corticospinal tract axons grew into the central portions of the bridges as shown by anterograde tracing of the corticospinal tract with BDA. The majority of the regenerated axons in the channels originated from local host neurons adjacent to the tissue bridges. In conclusion, we showed that growth factors increased survival of transplanted NSPCs whereas NgR enhanced axonal regeneration, but the combination did not have additive effects on functional recovery or regeneration.
Collapse
Affiliation(s)
- Xiaodong Guo
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
- Toronto Western Research Institute, Toronto Western Hospital and University of Toronto, Toronto, Ontario, Canada
| | - Tasneem Zahir
- Department of Chemical Engineering & Applied Chemistry, Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Andrea Mothe
- Toronto Western Research Institute, Toronto Western Hospital and University of Toronto, Toronto, Ontario, Canada
| | - Molly S. Shoichet
- Department of Chemical Engineering & Applied Chemistry, Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Cindi M. Morshead
- Department of Surgery and Institute of Medical Sciences, Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Yusuke Katayama
- Department of Chemical Engineering & Applied Chemistry, Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Charles H. Tator
- Toronto Western Research Institute, Toronto Western Hospital and University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
42
|
Kang ES, Ha KY, Kim YH. Fate of transplanted bone marrow derived mesenchymal stem cells following spinal cord injury in rats by transplantation routes. J Korean Med Sci 2012; 27:586-93. [PMID: 22690088 PMCID: PMC3369443 DOI: 10.3346/jkms.2012.27.6.586] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Accepted: 03/13/2012] [Indexed: 12/22/2022] Open
Abstract
This research was performed to investigate the differences of the transplanted cells' survival and differentiation, and its efficacy according to the delivery routes following spinal cord injury. Allogenic mesenchymal stem cells (MSCs) were transplanted intravenously (IV group) or intralesionally (IL group) at post-injury 1 day in rats. Behavioral improvement, engraftment and differentiation of the transplanted cells and the expression of neurotrophic factors of the transplanted groups were analyzed and compared with those of the control group. At 6 weeks post-injury, the mean BBB motor scales in the control, IV and IL groups were 6.5 ± 1.8, 11.1 ± 2.1, and 8.5 ± 2.8, respectively. Regardless of the delivery route, the MSCs transplantation following spinal cord injuries presented better behavioral improvement. The differentiations of the engrafted cells were different according to the delivery routes. The engrafted cells predominantly differentiated into astrocytes in the IV group and on the other hand, engrafted cells of the IL group demonstrated relatively even neural and glial differentiation. The expressions of neuronal growth factor were significantly higher in the IL group (mean relative optical density, 2.4 ± 0.15) than those in the control (2.16 ± 0.04) or IV group (1.7 ± 0.23). Transplantation of MSCs in the early stage of spinal cord injury gives a significant clinical improvement. However, the fate of the transplanted MSCs and expression of neuronal growth factors are different along the transplantation route.
Collapse
Affiliation(s)
- Eun-Sun Kang
- Department of Orthopedic Surgery, Seoul St. Mary's Hosptial, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Kee-Yong Ha
- Department of Orthopedic Surgery, Seoul St. Mary's Hosptial, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Young-Hoon Kim
- Department of Orthopedic Surgery, Seoul St. Mary's Hosptial, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
43
|
Hwang DH, Kim HM, Kang YM, Joo IS, Cho CS, Yoon BW, Kim SU, Kim BG. Combination of Multifaceted Strategies to Maximize the Therapeutic Benefits of Neural Stem Cell Transplantation for Spinal Cord Repair. Cell Transplant 2011; 20:1361-79. [DOI: 10.3727/096368910x557155] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Neural stem cells (NSCs) possess therapeutic potentials to reverse complex pathological processes following spinal cord injury (SCI), but many obstacles remain that could not be fully overcome by NSC transplantation alone. Combining complementary strategies might be required to advance NSC-based treatments to the clinical stage. The present study was undertaken to examine whether combination of NSCs, polymer scaffolds, neurotrophin-3 (NT3), and chondroitinase, which cleaves chondroitin sulfate proteoglycans at the interface between spinal cord and implanted scaffold, could provide additive therapeutic benefits. In a rat hemisection model, poly(e-caprolactone) (PCL) was used as a bridging scaffold and as a vehicle for NSC delivery. The PCL scaffolds seeded with F3 NSCs or NT3 overexpressing F3 cells (F3.NT3) were implanted into hemisected cavities. F3.NT3 showed better survival and migration, and more frequently differentiated into neurons and oligodendrocytes than F3 cells. Animals with PCL scaffold containing F3.NT3 cells showed the best locomotor recovery, and motor evoked potentials (MEPs) following transcranial magnetic stimulation were recorded only in PCL-F3.NT3 group in contralateral, but not ipsilateral, hindlimbs. Implantation of PCL scaffold with F3.NT3 cells increased NT3 levels, promoted neuroplasticity, and enhanced remyelination of contralateral white matter. Combining chondroitinase treatment after PCL-F3.NT3 implantation further enhanced cell migration and promoted axonal remodeling, and this was accompanied by augmented locomotor recovery and restoration of MEPs in ipsilateral hindlimbs. We demonstrate that combining multifaceted strategies can maximize the therapeutic benefits of NSC transplantation for SCI. Our results may have important clinical implications for the design of future NSC-based strategies.
Collapse
Affiliation(s)
- Dong H. Hwang
- Brain Disease Research Center, Institute of Medical Sciences, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Hyuk M. Kim
- Brain Disease Research Center, Institute of Medical Sciences, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Young M. Kang
- Brain Disease Research Center, Institute of Medical Sciences, Ajou University School of Medicine, Suwon, Republic of Korea
| | - In S. Joo
- Department of Neurology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Chong-Su Cho
- Department of Agricultural Biotechnology, Seoul National University, Seoul, Republic of Korea
| | - Byung-Woo Yoon
- Departments of Neurology and Neuroscience Research Center, Clinical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Seung U. Kim
- Department of Neurology, University of British Columbia, Vancouver, BC, Canada
- Medical Research Institute, Chungang University School of Medicine, Seoul, Republic of Korea
| | - Byung G. Kim
- Brain Disease Research Center, Institute of Medical Sciences, Ajou University School of Medicine, Suwon, Republic of Korea
- Department of Neurology, Ajou University School of Medicine, Suwon, Republic of Korea
| |
Collapse
|
44
|
Inoue H, Takenaga M, Ohta Y, Tomioka M, Watabe YI, Aihara M, Kumagai N. Improvement of hind-limb paralysis following traumatic spinal cord injury in rats by grafting normal human keratinocytes: new cell-therapy strategy for nerve regeneration. J Artif Organs 2011; 14:375-80. [DOI: 10.1007/s10047-011-0598-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2011] [Accepted: 07/28/2011] [Indexed: 12/28/2022]
|
45
|
Hefferan MP, Johe K, Hazel T, Feldman EL, Lunn JS, Marsala M. Optimization of immunosuppressive therapy for spinal grafting of human spinal stem cells in a rat model of ALS. Cell Transplant 2011; 20:1153-61. [PMID: 21669047 DOI: 10.3727/096368910x564553] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Previous rodent studies employing monotherapy or combined immunosuppressive regimens have demonstrated a variable degree of spinal xenograft survival in several spinal neurodegenerative models including spinal ischemia, trauma, or amyotrophic lateral sclerosis (ALS). Accordingly, the characterization of optimal immunosuppressive protocols for the specific neurodegenerative model is critical to ensure reliable assessment of potential long-term therapeutic effects associated with cell replacement. In the present study we characterized the survival of human spinal stem cells when grafted into the lumbar spinal cords of a rodent model of ALS, SOD1 (G93A) male and female rats (60-67 days old). Four different immunosuppressive protocols were studied: i) FK506 (q12h); ii) FK506 (qd) + mycophenolate (PO; q12h, up to 7 days postop); iii) FK506 (qd) + mycophenolate (IP; q12h, up to 7 days postop); and iv) FK506 (qd) + mycophenolate (IP; qd, up to 7 days postop). Three weeks after cell grafting the number of surviving human cells was then systematically assessed. The highest density of grafted cells was seen in animals treated with FK506 (qd) and mycophenolate (IP; qd; an average 915 ± 95 grafted cells per spinal cord section). The majority of hNUMA-positive cells colocalized with doublecortin (DCX) immunoreactivity. DCX-positive neurons showed extensive axodendritic sprouting toward surrounding host neurons. In addition, migrating grafted cells were identified up to 500 μm from the graft. In animals treated with FK506 (q12h), FK506 (qd) + mycophenolate (PO; q12h) or FK506 (qd) + mycophenolate (IP; q12h), 11.8 ± 3.4%, 61.2 ± 7.8%, and 99.4 ± 8.9% [expressed as percent of the FK506 (qd) and mycophenolate (IP; qd)] cell survival was seen, respectively. In contrast to animals treated with a combination of FK506 + mycophenolate, robust CD4/8 immunoreactivity was identified in the vicinity of the injection tract in animals treated with FK506 only. These data suggest that a combined, systemically delivered immunosuppression regimen including FK506 and mycophenolate can significantly improve survival of human spinal stem cells after intraspinal transplantation in SOD1 (G93A) rats.
Collapse
Affiliation(s)
- Michael P Hefferan
- Neuroregeneration Laboratory, Department of Anesthesiology, University of California-San Diego, La Jolla, CA 92093-0695, USA.
| | | | | | | | | | | |
Collapse
|
46
|
Neural stem cells modified by a hypoxia-inducible VEGF gene expression system improve cell viability under hypoxic conditions and spinal cord injury. Spine (Phila Pa 1976) 2011; 36:857-64. [PMID: 21192293 DOI: 10.1097/brs.0b013e3181e7f34b] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
STUDY DESIGN An in vitro neural hypoxia model and rat spinal cord injury (SCI) model were used to assess the regulation of therapeutic vascular endothelial growth factor (VEGF) gene expression in mouse neural stem cells (mNSCs) by the EPO (erythropoietin) enhancer or RTP801 promoter. OBJECTIVE To increase VEGF gene expression in mNSCs under hypoxic conditions in SCI lesions but avoid unwanted overexpression of VEGF in normal sites, we developed a hypoxia-inducible gene expression system consisting of the EPO enhancer and RTP801 promoter fused to VEGF or the luciferase gene, then transfected into mNSCs. SUMMARY OF BACKGROUND DATA On the basis of the ischemic response in the injured area, poor cell survival at the transplantation site is a consistent problem with NSC transplantation after SCI. Although VEGF directly protects neurons and enhances neurite outgrowth, uncontrolled overexpression of VEGF in uninjured tissue may cause serious adverse effects. To effectively improve NSC survival in ischemic sites after transplantation, we evaluated mNSCs modified by a hypoxia-inducible VEGF gene expression system in an SCI model. METHODS Hypoxia-inducible luciferase or VEGF plasmids were constructed using the EPO enhancer or RTP801 promoter. The effect of these systems on targeted gene expression and cell viability was evaluated in mNSCs in both hypoxic in vitro injury and a rat SCI model in vivo. RESULTS The gene expression system containing the EPO enhancer or RTP801 promoter significantly increased the expression of the luciferase reporter gene and therapeutic VEGF gene under hypoxic conditions. The Epo-SV-VEGF plasmid transfection group had significantly fewer apoptotic cells in vitro. This system also augmented cell viability in the in vivo SCI model. CONCLUSION These results strongly suggest the potential utility of mNSCs modified by a hypoxia-inducible VEGF gene expression system in the development of effective stem cell transplantation protocols in SCI.
Collapse
|
47
|
Urdziková L, Likavčanová-Mašínová K, Vaněček V, Růžička J, Sedý J, Syková E, Jendelová P. Flt3 ligand synergizes with granulocyte-colony-stimulating factor in bone marrow mobilization to improve functional outcome after spinal cord injury in the rat. Cytotherapy 2011; 13:1090-104. [PMID: 21539498 DOI: 10.3109/14653249.2011.575355] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND AIMS The effect of granulocyte-colony-stimulating factor (G-CSF) and/or the cytokine fms-like thyrosin kinase 3 (Flt3) ligand on functional outcome and tissue regeneration was studied in a rat model of spinal cord injury (SCI). METHODS Rats with a balloon-induced compression lesion were injected with G-CSF and/or Flt3 ligand to mobilize bone marrow cells. Behavioral tests (Basso-Beattie-Bresnahan and plantar test), blood counts, morphometric evaluation of the white and gray matter, and histology were performed 5 weeks after SCI. RESULTS The mobilization of bone marrow cells by G-CSF, Flt3 ligand and their combination improved the motor and sensory performance of rats with SCI, reduced glial scarring, increased axonal sprouting and spared white and gray matter in the lesion. The best results were obtained with a combination of G-CSF and Flt3. G-CSF alone or in combination with Flt3 ligand significantly increased the number of white blood cells, but not red blood cells or hemoglobin content, during and after the time-course of bone marrow stimulation. The combination of factors led to infiltration of the lesion by CD11b(+) cells. CONCLUSIONS The observed improvement in behavioral and morphologic parameters and tissue regeneration in animals with SCI treated with a combination of both factors could be associated with a prolonged time-course of mobilization of bone marrow cells. The intravenous administration of G-CSF and/or Flt3 ligand represents a safe and effective treatment modality for SCI.
Collapse
Affiliation(s)
- Lucia Urdziková
- Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | | | | | | | | | | | | |
Collapse
|
48
|
Bauchet L, Lonjon N, Perrin FE, Gilbert C, Privat A, Fattal C. Strategies for spinal cord repair after injury: a review of the literature and information. Ann Phys Rehabil Med 2011; 52:330-51. [PMID: 19886026 DOI: 10.1016/j.annrmp.2008.10.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Thanks to the Internet, we can now have access to more information about spinal cord repair. Spinal cord injured (SCI) patients request more information and hospitals offer specific spinal cord repair medical consultations. OBJECTIVE Provide practical and relevant elements to physicians and other healthcare professionals involved in the care of SCI patients in order to provide adequate answers to their questions. METHOD Our literature review was based on English and French publications indexed in PubMed and the main Internet websites dedicated to spinal cord repair. RESULTS A wide array of research possibilities including notions of anatomy, physiology, biology, anatomopathology and spinal cord imaging is available for the global care of the SCI patient. Prevention and repair strategies (regeneration, transplant, stem cells, gene therapy, biomaterials, using sublesional uninjured spinal tissue, electrical stimulation, brain/computer interface, etc.) for the injured spinal cord are under development. It is necessary to detail the studies conducted and define the limits of these new strategies and benchmark them to the realistic medical and rehabilitation care available to these patients. CONCLUSION Research is quickly progressing and clinical trials will be developed in the near future. They will have to answer to strict methodological and ethical guidelines. They will first be designed for a small number of patients. The results will probably be fragmented and progress will be made through different successive steps.
Collapse
Affiliation(s)
- L Bauchet
- Centre mutualiste neurologique Propara, 34195 Montpellier, France.
| | | | | | | | | | | |
Collapse
|
49
|
Park DH, Lee JH, Borlongan CV, Sanberg PR, Chung YG, Cho TH. Transplantation of umbilical cord blood stem cells for treating spinal cord injury. Stem Cell Rev Rep 2011; 7:181-94. [PMID: 20532836 DOI: 10.1007/s12015-010-9163-0] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Spinal cord injury (SCI) develops primary and secondary damage to neural tissue and this often results in permanent disability of the motor and sensory functions. However, there is currently no effective treatment except methylprednisolone, and the use of methylprednisolone has also been questioned due to its moderate efficacy and the drug's downside. Regenerative medicine has remarkably developed since the discovery of stem cells, and many studies have suggested the potential of cell-based therapies for neural injury. Especially, the therapeutic potential of human umbilical cord blood cells (hUCB cells) for intractable neurological disorders has been demonstrated using in vitro and vivo models. The hUCB cells are immune naïve and they are able to differentiate into other phenotypes, including the neural lineage. Their ability to produce several neurotropic factors and to modulate immune and inflammatory reactions has also been noted. Recent evidence has emerged suggesting alternative pathways of graft-mediated neural repair that involve neurotrophic effects. These effects are caused by the release of various growth factors that promote cell survival, angiogenesis and anti-inflammation, and this is all aside from a cell replacement mechanism. In this review, we present the recent findings on the stemness properties and the therapeutic potential of hUCB as a safe, feasible and effective cellular source for transplantation in SCI. These multifaceted protective and restorative effects from hUCB grafts may be interdependent and they act in harmony to promote therapeutic benefits for SCI. Nevertheless, clinical studies with hUCB are still rare because of the concerns about safety and efficiency. Among these concerns, the major histocompatibility in allogeneic transplantation is an important issue to be addressed in future clinical trials for treating SCI.
Collapse
Affiliation(s)
- Dong-Hyuk Park
- Department of Neurosurgery, Korea University Medical Center, Anam Hospital, Korea University College of Medicine, #126, 5-GA, Anam-Dong, Sungbuk-Ku, Seoul 136-705, Korea.
| | | | | | | | | | | |
Collapse
|
50
|
Jia X, Kowalski RG, Sciubba DM, Geocadin RG. Critical care of traumatic spinal cord injury. J Intensive Care Med 2011; 28:12-23. [PMID: 21482574 DOI: 10.1177/0885066611403270] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Approximately 11 000 people suffer traumatic spinal cord injury (TSCI) in the United States, each year. TSCI incidences vary from 13.1 to 52.2 per million people and the mortality rates ranged from 3.1 to 17.5 per million people. This review examines the critical care of TSCI. The discussion will focus on primary and secondary mechanisms of injury, spine stabilization and immobilization, surgery, intensive care management, airway and respiratory management, cardiovascular complication management, venous thromboembolism, nutrition and glucose control, infection management, pressure ulcers and early rehabilitation, pharmacologic cord protection, and evolving treatment options including the use of pluripotent stem cells and hypothermia.
Collapse
Affiliation(s)
- Xiaofeng Jia
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | | | | | | |
Collapse
|