1
|
Farid MF, S Abouelela Y, Rizk H. Stem cell treatment trials of spinal cord injuries in animals. Auton Neurosci 2022; 238:102932. [PMID: 35016045 DOI: 10.1016/j.autneu.2021.102932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 11/01/2021] [Accepted: 12/23/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND Spinal cord injury (SCI) is a serious neurological spinal cord damage that resulted in the loss of temporary or permanent function. However, there are even now no effective therapies for it. So, a new medical promising therapeutic hotspot over the previous decades appeared which was (Stem cell (SC) cure of SCI). Otherwise, animal models are considered in preclinical research as a model for humans to trial a potential new treatment. METHODOLOGY Following articles were saved from different databases (PubMed, Google scholar, Egyptian knowledge bank, Elsevier, Medline, Embase, ProQuest, BMC) on the last two decades, and data were obtained then analyzed. RESULTS This review discusses the type and grading of SCI. As well as different types of stem cells therapy for SCI, including mesenchymal stem cells (MSCs), neural stem cells (NSCs), hematopoietic stem cells (HSCs), induced pluripotent stem cells (iPSCs), and embryonic stem cells (ESCs). The review focuses on the transplantation pathways, clinical evaluation, and clinical signs of different types of SC on different animal models which are summarized in tables to give an easy to reach. CONCLUSION Pharmacological and physiotherapy have limited regenerative power in comparison with stem cells medication in the treatment of SCI. Among several sources of cell therapies, mesenchymal stromal/stem cell (MSC) one is being progressively developed as a trusted important energetic way to repair and regenerate. Finally, a wide-ranged animal models have been condensed that helped in human clinical trial therapies.
Collapse
Affiliation(s)
- Mariam F Farid
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt
| | - Yara S Abouelela
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt.
| | - Hamdy Rizk
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt
| |
Collapse
|
2
|
Richard SA, Sackey M. Elucidating the Pivotal Neuroimmunomodulation of Stem Cells in Spinal Cord Injury Repair. Stem Cells Int 2021; 2021:9230866. [PMID: 34341666 PMCID: PMC8325586 DOI: 10.1155/2021/9230866] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 07/03/2021] [Accepted: 07/17/2021] [Indexed: 12/11/2022] Open
Abstract
Spinal cord injury (SCI) is a distressing incident with abrupt onset of the motor as well as sensory dysfunction, and most often, the injury occurs as result of high-energy or velocity accidents as well as contact sports and falls in the elderly. The key challenges associated with nerve repair are the lack of self-repair as well as neurotrophic factors and primary and secondary neuronal apoptosis, as well as factors that prevent the regeneration of axons locally. Neurons that survive the initial traumatic damage may be lost due to pathogenic activities like neuroinflammation and apoptosis. Implanted stem cells are capable of differentiating into neural cells that replace injured cells as well as offer local neurotrophic factors that aid neuroprotection, immunomodulation, axonal sprouting, axonal regeneration, and remyelination. At the microenvironment of SCI, stem cells are capable of producing growth factors like brain-derived neurotrophic factor and nerve growth factor which triggers neuronal survival as well as axonal regrowth. Although stem cells have proven to be of therapeutic value in SCI, the major disadvantage of some of the cell types is the risk for tumorigenicity due to the contamination of undifferentiated cells prior to transplantation. Local administration of stem cells via either direct cellular injection into the spinal cord parenchyma or intrathecal administration into the subarachnoid space is currently the best transplantation modality for stem cells during SCI.
Collapse
Affiliation(s)
- Seidu A. Richard
- Department of Medicine, Princefield University, P.O. Box MA128, Ho, Ghana
| | - Marian Sackey
- Department of Pharmacy, Ho Teaching Hospital, P.O. Box MA-374, Ho, Ghana
| |
Collapse
|
3
|
Bonilla C, Zurita M. Cell-Based Therapies for Traumatic Brain Injury: Therapeutic Treatments and Clinical Trials. Biomedicines 2021; 9:biomedicines9060669. [PMID: 34200905 PMCID: PMC8230536 DOI: 10.3390/biomedicines9060669] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/09/2021] [Accepted: 06/09/2021] [Indexed: 02/07/2023] Open
Abstract
Traumatic brain injury (TBI) represents physical damage to the brain tissue that induces transitory or permanent neurological disabilities. TBI contributes to 50% of all trauma deaths, with many enduring long-term consequences and significant medical and rehabilitation costs. There is currently no therapy to reverse the effects associated with TBI. An increasing amount of research has been undertaken regarding the use of different stem cells (SCs) to treat the consequences of brain damage. Neural stem cells (NSCs) (adult and embryonic) and mesenchymal stromal cells (MSCs) have shown efficacy in pre-clinical models of TBI and in their introduction to clinical research. The purpose of this review is to provide an overview of TBI and the state of clinical trials aimed at evaluating the use of stem cell-based therapies in TBI. The primary aim of these studies is to investigate the safety and efficacy of the use of SCs to treat this disease. Although an increasing number of studies are being carried out, few results are currently available. In addition, we present our research regarding the use of cell therapy in TBI. There is still a significant lack of understanding regarding the cell therapy mechanisms for the treatment of TBI. Thus, future studies are needed to evaluate the feasibility of the transplantation of SCs in TBI.
Collapse
Affiliation(s)
- Celia Bonilla
- Cell Therapy Unit, Puerta de Hierro Hospital, 28222 Majadahonda, Madrid, Spain
- Correspondence: ; Tel.: +34-91-191-7879
| | - Mercedes Zurita
- Cell Therapy Unit Responsable, Puerta de Hierro Hospital, 28222 Majadahonda, Madrid, Spain;
| |
Collapse
|
4
|
Krull AA, Setter DO, Gendron TF, Hrstka SCL, Polzin MJ, Hart J, Dudakovic A, Madigan NN, Dietz AB, Windebank AJ, van Wijnen AJ, Staff NP. Alterations of mesenchymal stromal cells in cerebrospinal fluid: insights from transcriptomics and an ALS clinical trial. Stem Cell Res Ther 2021; 12:187. [PMID: 33736701 PMCID: PMC7977179 DOI: 10.1186/s13287-021-02241-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 02/24/2021] [Indexed: 12/12/2022] Open
Abstract
Background Mesenchymal stromal cells (MSCs) have been studied with increasing intensity as clinicians and researchers strive to understand the ability of MSCs to modulate disease progression and promote tissue regeneration. As MSCs are used for diverse applications, it is important to appreciate how specific physiological environments may stimulate changes that alter the phenotype of the cells. One need for neuroregenerative applications is to characterize the spectrum of MSC responses to the cerebrospinal fluid (CSF) environment after their injection into the intrathecal space. Mechanistic understanding of cellular biology in response to the CSF environment may predict the ability of MSCs to promote injury repair or provide neuroprotection in neurodegenerative diseases. Methods In this study, we characterized changes in morphology, metabolism, and gene expression occurring in human adipose-derived MSCs cultured in human (hCSF) or artificial CSF (aCSF) as well as examined relevant protein levels in the CSF of subjects treated with MSCs for amyotrophic lateral sclerosis (ALS). Results Our results demonstrated that, under intrathecal-like conditions, MSCs retained their morphology, though they became quiescent. Large-scale transcriptomic analysis of MSCs revealed a distinct gene expression profile for cells cultured in aCSF. The aCSF culture environment induced expression of genes related to angiogenesis and immunomodulation. In addition, MSCs in aCSF expressed genes encoding nutritional growth factors to expression levels at or above those of control cells. Furthermore, we observed a dose-dependent increase in growth factors and immunomodulatory cytokines in CSF from subjects with ALS treated intrathecally with autologous MSCs. Conclusions Overall, our results suggest that MSCs injected into the intrathecal space in ongoing clinical trials remain viable and may provide a therapeutic benefit to patients. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02241-9.
Collapse
Affiliation(s)
- Ashley A Krull
- Department of Neurology, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Deborah O Setter
- Department of Neurology, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Tania F Gendron
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Sybil C L Hrstka
- Department of Neurology, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Michael J Polzin
- Department of Neurology, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Joseph Hart
- Department of Neurology, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Amel Dudakovic
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, 55905, USA
| | - Nicolas N Madigan
- Department of Neurology, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Allan B Dietz
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Anthony J Windebank
- Department of Neurology, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA
| | - Andre J van Wijnen
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, 55905, USA
| | - Nathan P Staff
- Department of Neurology, Mayo Clinic, 200 First St. SW, Rochester, MN, 55905, USA.
| |
Collapse
|
5
|
Yoo SH, Lee SH, Lee S, Park JH, Lee S, Jin H, Park HJ. The effect of human mesenchymal stem cell injection on pain behavior in chronic post-ischemia pain mice. Korean J Pain 2020; 33:23-29. [PMID: 31888314 PMCID: PMC6944374 DOI: 10.3344/kjp.2020.33.1.23] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 10/29/2019] [Accepted: 11/01/2019] [Indexed: 12/30/2022] Open
Abstract
Background Neuropathic pain (NP) is considered a clinically incurable condition despite various treatment options due to its diverse causes and complicated disease mechanisms. Since the early 2000s, multipotent human mesenchymal stem cells (hMSCs) have been used in the treatment of NP in animal models. However, the effects of hMSC injections have not been studied in chronic post-ischemia pain (CPIP) mice models. Here, we investigated whether intrathecal (IT) and intrapaw (IP) injections of hMSCs can reduce mechanical allodynia in CPIP model mice. Methods Seventeen CPIP C57/BL6 mice were selected and randomized into four groups: IT sham (n = 4), IT stem (n = 5), IP sham (n = 4), and IP stem (n = 4). Mice in the IT sham and IT stem groups received an injection of 5 μL saline and 2 × 104 hMSCs, respectively, while mice in the IP sham and IP stem groups received an injection of 5 μL saline and 2 × 105 hMSCs, respectively. Mechanical allodynia was assessed using von Frey filaments from pre-injection to 30 days post-injection. Glial fibrillary acidic protein (GFAP) expression in the spinal cord and dorsal root ganglia were also evaluated. Results IT and IP injections of hMSCs improved mechanical allodynia. GFAP expression was decreased on day 25 post-injection compared with the sham group. Injections of hMSCs improved allodynia and GFAP expression was decreased compared with the sham group. Conclusions These results suggested that hMSCs may be also another treatment modality in NP model by ischemia-reperfusion.
Collapse
Affiliation(s)
- Sie Hyeon Yoo
- Department of Anesthesiology and Pain Medicine, Soonchunhyang University Cheonan Hospital, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Sung Hyun Lee
- Department of Anesthesiology and Pain Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Seunghwan Lee
- Department of Anesthesiology and Pain Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jae Hong Park
- Department of Anesthesiology and Pain Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Seunghyeon Lee
- Department of Anesthesiology and Pain Medicine, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Korea
| | - Heecheol Jin
- Department of Anesthesiology and Pain Medicine, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Korea
| | - Hue Jung Park
- Department of Anesthesiology and Pain Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
6
|
Upregulation of UBAP2L in Bone Marrow Mesenchymal Stem Cells Promotes Functional Recovery in Rats with Spinal Cord Injury. Curr Med Sci 2018; 38:1081-1089. [PMID: 30536073 DOI: 10.1007/s11596-018-1987-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 06/12/2018] [Indexed: 02/07/2023]
Abstract
Post-translational modifications of cellular proteins with ubiquitin or ubiquitin-like proteins regulate many cellular processes, such as cell proliferation, differentiation, apoptosis, signal transduction, intercellular immune recognition, inflammatory response, stress response, and DNA repair. Nice4/UBAP2L is an important member in the family of ubiquitin-like proteins, and its biological function remains unknown. This study aimed to investigate the effect of UBAP2L on spinal cord injury (SCI). At first, rat bone marrow mesenchymal stem cells (BMSCs) were infected with adeno-associated virus to induce over-expression of Nice4. Subsequently, the infected BMSCs were transplanted into rats suffering from semi-sectioned SCI. The results showed that the over-expression of Nice4 significantly promoted the proliferation and differentiation of BMSCs. In addition, the transplantation of infected BMSCs into the injured area of SCI rats improved the function repair of SCI. Importantly, the immunohistochemical and hematoxylin-eosin staining and RT-PCR results showed that the number of neuronal cells, oligodendrocytes, and astrocytes was significantly increased in the injured area, along with significantly upregulated expression of cyclin D1 and p38 mitogen-activated protein kinase (MAPK). Meanwhile, the expression of caspase 3 protein was significantly down-regulated. In conclusion, the over-expression of Nice4 gene can promote the functional recovery in SCI rats by promoting cell proliferation and inhibiting apoptosis. The results of this study indicate an alternative option for the clinical treatment of SCI.
Collapse
|
7
|
Platelet-rich plasma-derived scaffolds increase the benefit of delayed mesenchymal stromal cell therapy after severe traumatic brain injury. Cytotherapy 2018; 20:314-321. [PMID: 29306567 DOI: 10.1016/j.jcyt.2017.11.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 09/11/2017] [Accepted: 11/30/2017] [Indexed: 12/28/2022]
Abstract
BACKGROUND Cell therapy using mesenchymal stromal cells (MSCs) offers new perspectives in the treatment of traumatic brain injury (TBI). The aim of the present study was to assess the impact of platelet-rich plasma scaffolds (PRPS) as support of MSCs in a delayed phase after severe TBI in rats. METHODS TBI was produced by weight-drop impact to the right cerebral hemisphere. Two months after TBI, four experimental groups were established; saline, PRPS, MSCs in saline, or MSCs in PRPS was transplanted into the area of brain lesion through a small hole. All groups were evaluated in the course of the following 12 months after therapy and the animals were then humanely killed. RESULTS Our results showed that a greater functional improvement was obtained after the administration of MSCs in PRPS compared with the other experimental groups. DISCUSSION PRPS enhanced the benefit of cell therapy with MSCs to treat chronic brain damage in rats that suffered a severe TBI. The present findings suggest that the use of intralesional MSCs supported in PRPS may be a strategy of tissue engineering for patients with established neurological severe dysfunction after a TBI.
Collapse
|
8
|
Pathophysiology, mechanisms and applications of mesenchymal stem cells for the treatment of spinal cord injury. Biomed Pharmacother 2017; 91:693-706. [DOI: 10.1016/j.biopha.2017.04.126] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 04/25/2017] [Accepted: 04/30/2017] [Indexed: 02/06/2023] Open
|
9
|
Nandoe Tewarie RDS, Nandoe RDS, Hurtado A, Levi ADO, Grotenhuis JA, Grotenhuis A, Oudega M. Bone Marrow Stromal Cells for Repair of the Spinal Cord: Towards Clinical Application. Cell Transplant 2017; 15:563-77. [PMID: 17176609 DOI: 10.3727/000000006783981602] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Stem cells have been recognized and intensively studied for their potential use in restorative approaches for degenerative diseases and traumatic injuries. In the central nervous system (CNS), stem cell-based strategies have been proposed to replace lost neurons in degenerative diseases such as Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis (Lou Gehrig's disease), or to replace lost oligodendrocytes in demyelinating diseases such as multiple sclerosis. Stem cells have also been implicated in repair of the adult spinal cord. An impact to the spinal cord results in immediate damage to tissue including blood vessels, causing loss of neurons, astrocytes, and oligodendrocytes. In time, more tissue nearby or away from the injury site is lost due to secondary injury. In case of relatively minor damage to the cord some return of function can be observed, but in most cases the neurological loss is permanent. This review will focus on in vitro and in vivo studies on the use of bone marrow stromal cells (BMSCs), a heterogeneous cell population that includes mesenchymal stem cells, for repair of the spinal cord in experimental injury models and their potential for human application. To optimally benefit from BMSCs for repair of the spinal cord it is imperative to develop in vitro techniques that will generate the desired cell type and/or a large enough number for in vivo transplantation approaches. We will also assess the potential and possible pitfalls for use of BMSCs in humans and ongoing clinical trials.
Collapse
Affiliation(s)
- Rishi D S Nandoe Tewarie
- The Miami Project to Cure Paralysis, University of Miami, School of Medicine, Miami, FL 33136, USA
| | | | | | | | | | | | | |
Collapse
|
10
|
Bastidas J, Athauda G, De La Cruz G, Chan WM, Golshani R, Berrocal Y, Henao M, Lalwani A, Mannoji C, Assi M, Otero PA, Khan A, Marcillo AE, Norenberg M, Levi AD, Wood PM, Guest JD, Dietrich WD, Bartlett Bunge M, Pearse DD. Human Schwann cells exhibit long-term cell survival, are not tumorigenic and promote repair when transplanted into the contused spinal cord. Glia 2017; 65:1278-1301. [PMID: 28543541 DOI: 10.1002/glia.23161] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Revised: 04/07/2017] [Accepted: 04/10/2017] [Indexed: 12/26/2022]
Abstract
The transplantation of rodent Schwann cells (SCs) provides anatomical and functional restitution in a variety of spinal cord injury (SCI) models, supporting the recent translation of SCs to phase 1 clinical trials for human SCI. Whereas human (Hu)SCs have been examined experimentally in a complete SCI transection paradigm, to date the reported behavior of SCs when transplanted after a clinically relevant contusive SCI has been restricted to the use of rodent SCs. Here, in a xenotransplant, contusive SCI paradigm, the survival, biodistribution, proliferation and tumorgenicity as well as host responses to HuSCs, cultured according to a protocol analogous to that developed for clinical application, were investigated. HuSCs persisted within the contused nude rat spinal cord through 6 months after transplantation (longest time examined), exhibited low cell proliferation, displayed no evidence of tumorigenicity and showed a restricted biodistribution to the lesion. Neuropathological examination of the CNS revealed no adverse effects of HuSCs. Animals exhibiting higher numbers of surviving HuSCs within the lesion showed greater volumes of preserved white matter and host rat SC and astrocyte ingress as well as axon ingrowth and myelination. These results demonstrate the safety of HuSCs when employed in a clinically relevant experimental SCI paradigm. Further, signs of a potentially positive influence of HuSC transplants on host tissue pathology were observed. These findings show that HuSCs exhibit a favorable toxicity profile for up to 6 months after transplantation into the contused rat spinal cord, an important outcome for FDA consideration of their use in human clinical trials.
Collapse
Affiliation(s)
- Johana Bastidas
- The Miami Project to Cure Paralysis, The Department of Neurological Surgery, The University of Miami Miller School of Medicine, Miami, Florida, 33136
| | - Gagani Athauda
- The Department of Cellular Biology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, 33199.,The Department of Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, 33199
| | - Gabriela De La Cruz
- Translational Pathology Laboratory, Lineberger Comprehensive Cancer Center, Department of Pathology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, 27599
| | - Wai-Man Chan
- The Miami Project to Cure Paralysis, The Department of Neurological Surgery, The University of Miami Miller School of Medicine, Miami, Florida, 33136
| | - Roozbeh Golshani
- The Miami Project to Cure Paralysis, The Department of Neurological Surgery, The University of Miami Miller School of Medicine, Miami, Florida, 33136
| | - Yerko Berrocal
- The Department of Cellular Biology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, 33199.,The Department of Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, 33199
| | - Martha Henao
- The Miami Project to Cure Paralysis, The Department of Neurological Surgery, The University of Miami Miller School of Medicine, Miami, Florida, 33136
| | - Anil Lalwani
- The Miami Project to Cure Paralysis, The Department of Neurological Surgery, The University of Miami Miller School of Medicine, Miami, Florida, 33136
| | - Chikato Mannoji
- The Department of Orthopedic Surgery, Chiba University School of Medicine, Chiba, Japan
| | - Mazen Assi
- The Miami Project to Cure Paralysis, The Department of Neurological Surgery, The University of Miami Miller School of Medicine, Miami, Florida, 33136
| | - P Anthony Otero
- The Miami Project to Cure Paralysis, The Department of Neurological Surgery, The University of Miami Miller School of Medicine, Miami, Florida, 33136
| | - Aisha Khan
- The Miami Project to Cure Paralysis, The Department of Neurological Surgery, The University of Miami Miller School of Medicine, Miami, Florida, 33136
| | - Alexander E Marcillo
- The Miami Project to Cure Paralysis, The Department of Neurological Surgery, The University of Miami Miller School of Medicine, Miami, Florida, 33136
| | - Michael Norenberg
- The Department of Pathology, The University of Miami Miller School of Medicine, Miami, Florida, 33136
| | - Allan D Levi
- The Miami Project to Cure Paralysis, The Department of Neurological Surgery, The University of Miami Miller School of Medicine, Miami, Florida, 33136.,The Department of Neurological Surgery, The University of Miami Miller School of Medicine, Miami, Florida, 33136
| | - Patrick M Wood
- The Miami Project to Cure Paralysis, The Department of Neurological Surgery, The University of Miami Miller School of Medicine, Miami, Florida, 33136
| | - James D Guest
- The Miami Project to Cure Paralysis, The Department of Neurological Surgery, The University of Miami Miller School of Medicine, Miami, Florida, 33136.,The Department of Neurological Surgery, The University of Miami Miller School of Medicine, Miami, Florida, 33136
| | - W Dalton Dietrich
- The Miami Project to Cure Paralysis, The Department of Neurological Surgery, The University of Miami Miller School of Medicine, Miami, Florida, 33136.,The Department of Neurological Surgery, The University of Miami Miller School of Medicine, Miami, Florida, 33136.,The Department of Neurology, The University of Miami Miller School of Medicine, Miami, Florida, 33136.,The Neuroscience Program, The University of Miami Miller School of Medicine, Miami, Florida, 33136.,The Interdisciplinary Stem Cell Institute, The University of Miami Miller School of Medicine, Miami, Florida, 33136.,The Department of Cell Biology, The University of Miami Miller School of Medicine, Miami, Florida, 33136
| | - Mary Bartlett Bunge
- The Miami Project to Cure Paralysis, The Department of Neurological Surgery, The University of Miami Miller School of Medicine, Miami, Florida, 33136.,The Department of Neurological Surgery, The University of Miami Miller School of Medicine, Miami, Florida, 33136.,The Neuroscience Program, The University of Miami Miller School of Medicine, Miami, Florida, 33136.,The Interdisciplinary Stem Cell Institute, The University of Miami Miller School of Medicine, Miami, Florida, 33136.,The Department of Cell Biology, The University of Miami Miller School of Medicine, Miami, Florida, 33136
| | - Damien D Pearse
- The Miami Project to Cure Paralysis, The Department of Neurological Surgery, The University of Miami Miller School of Medicine, Miami, Florida, 33136.,The Department of Neurological Surgery, The University of Miami Miller School of Medicine, Miami, Florida, 33136.,The Neuroscience Program, The University of Miami Miller School of Medicine, Miami, Florida, 33136.,The Interdisciplinary Stem Cell Institute, The University of Miami Miller School of Medicine, Miami, Florida, 33136.,Bruce W. Carter Department of Veterans Affairs Medical Center, Miami, Florida, 33136
| |
Collapse
|
11
|
Vaquero J, Zurita M, Rico MA, Bonilla C, Aguayo C, Fernández C, Tapiador N, Sevilla M, Morejón C, Montilla J, Martínez F, Marín E, Bustamante S, Vázquez D, Carballido J, Rodríguez A, Martínez P, García C, Ovejero M, Fernández MV. Repeated subarachnoid administrations of autologous mesenchymal stromal cells supported in autologous plasma improve quality of life in patients suffering incomplete spinal cord injury. Cytotherapy 2017; 19:349-359. [PMID: 28089079 DOI: 10.1016/j.jcyt.2016.12.002] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Revised: 12/12/2016] [Accepted: 12/13/2016] [Indexed: 12/26/2022]
Abstract
BACKGROUND AIMS Cell therapy with mesenchymal stromal cells (MSCs) offers new hope for patients suffering from spinal cord injury (SCI). METHODS Ten patients with established incomplete SCI received four subarachnoid administrations of 30 × 106 autologous bone marrow MSCs, supported in autologous plasma, at months 1, 4, 7 and 10 of the study, and were followed until the month 12. Urodynamic, neurophysiological and neuroimaging studies were performed at months 6 and 12, and compared with basal studies. RESULTS Variable improvement was found in the patients of the series. All of them showed some degree of improvement in sensitivity and motor function. Sexual function improved in two of the eight male patients. Neuropathic pain was present in four patients before treatment; it disappeared in two of them and decreased in another. Clear improvement in bladder and bowel control were found in all patients suffering previous dysfunction. Before treatment, seven patients suffered spasms, and two improved. Before cell therapy, nine patients suffered variable degree of spasticity, and 3 of them showed clear decrease at the end of follow-up. At this time, nine patients showed infra-lesional electromyographic recordings suggesting active muscle reinnervation, and eight patients showed improvement in bladder compliance. After three administrations of MSCs, mean values of brain-derived neurotrophic factor, glial-derived neurotrophic factor, ciliary neurotrophic factor, and neurotrophin 3 and 4 showed slight increases compared with basal levels, but without statistically significant difference. CONCLUSIONS Administration of repeated doses of MSCs by subarachnoid route is a well-tolerated procedure that is able to achieve progressive and significant improvement in the quality of life of patients suffering incomplete SCI.
Collapse
Affiliation(s)
- Jesús Vaquero
- Neurosurgery Service, University Hospital Puerta de Hierro-Majadahonda, Autonomous University, Madrid, Spain; Neuroscience Research Unit, University Hospital Puerta de Hierro-Majadahonda, Autonomous University, Madrid, Spain.
| | - Mercedes Zurita
- Neuroscience Research Unit, University Hospital Puerta de Hierro-Majadahonda, Autonomous University, Madrid, Spain
| | - Miguel A Rico
- Neuroscience Research Unit, University Hospital Puerta de Hierro-Majadahonda, Autonomous University, Madrid, Spain
| | - Celia Bonilla
- Neuroscience Research Unit, University Hospital Puerta de Hierro-Majadahonda, Autonomous University, Madrid, Spain
| | - Concepción Aguayo
- Neuroscience Research Unit, University Hospital Puerta de Hierro-Majadahonda, Autonomous University, Madrid, Spain
| | - Cecilia Fernández
- Neurosurgery Service, University Hospital Puerta de Hierro-Majadahonda, Autonomous University, Madrid, Spain
| | - Noemí Tapiador
- Rehabilitation Service, University Hospital Puerta de Hierro-Majadahonda, Autonomous University, Madrid, Spain
| | - Marta Sevilla
- Rehabilitation Service, University Hospital Puerta de Hierro-Majadahonda, Autonomous University, Madrid, Spain
| | - Carlos Morejón
- Rehabilitation Service, University Hospital Puerta de Hierro-Majadahonda, Autonomous University, Madrid, Spain
| | - Jesús Montilla
- Rehabilitation Service, University Hospital Puerta de Hierro-Majadahonda, Autonomous University, Madrid, Spain
| | - Francisco Martínez
- Clinical Neurophysiology Service, University Hospital Puerta de Hierro-Majadahonda, Autonomous University, Madrid, Spain
| | - Esperanza Marín
- Clinical Neurophysiology Service, University Hospital Puerta de Hierro-Majadahonda, Autonomous University, Madrid, Spain
| | - Salvador Bustamante
- Urology Service, University Hospital Puerta de Hierro-Majadahonda, Autonomous University, Madrid, Spain
| | - David Vázquez
- Urology Service, University Hospital Puerta de Hierro-Majadahonda, Autonomous University, Madrid, Spain
| | - Joaquín Carballido
- Urology Service, University Hospital Puerta de Hierro-Majadahonda, Autonomous University, Madrid, Spain
| | - Alicia Rodríguez
- Neuroscience Research Unit, University Hospital Puerta de Hierro-Majadahonda, Autonomous University, Madrid, Spain
| | - Paula Martínez
- Neuroscience Research Unit, University Hospital Puerta de Hierro-Majadahonda, Autonomous University, Madrid, Spain
| | - Coral García
- Neuroimmunology Unit, University Hospital Puerta de Hierro-Majadahonda, Autonomous University, Madrid, Spain
| | | | - Marta V Fernández
- Neuroscience Research Unit, University Hospital Puerta de Hierro-Majadahonda, Autonomous University, Madrid, Spain
| | | |
Collapse
|
12
|
Vaquero J, Zurita M, Rico MA, Bonilla C, Aguayo C, Montilla J, Bustamante S, Carballido J, Marin E, Martinez F, Parajon A, Fernandez C, Reina LD. An approach to personalized cell therapy in chronic complete paraplegia: The Puerta de Hierro phase I/II clinical trial. Cytotherapy 2016; 18:1025-1036. [PMID: 27311799 DOI: 10.1016/j.jcyt.2016.05.003] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 04/21/2016] [Accepted: 05/08/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND AIMS Cell transplantation in patients suffering spinal cord injury (SCI) is in its initial stages, but currently there is confusion about the results because of the disparity in the techniques used, the route of administration, and the criteria for selecting patients. METHODS We conducted a clinical trial involving 12 patients with complete and chronic paraplegia (average time of chronicity, 13.86 years; SD, 9.36). The characteristics of SCI in magnetic resonance imaging (MRI) were evaluated for a personalized local administration of expanded autologous bone marrow mesenchymal stromal cells (MSCs) supported in autologous plasma, with the number of MSCs ranging from 100 × 10(6) to 230 × 10(6). An additional 30 × 10(6) MSCs were administered 3 months later by lumbar puncture into the subarachnoid space. Outcomes were evaluated at 3, 6, 9 and 12 months after surgery through clinical, urodynamic, neurophysiological and neuroimaging studies. RESULTS Cell transplantation is a safe procedure. All patients experienced improvement, primarily in sensitivity and sphincter control. Infralesional motor activity, according to clinical and neurophysiological studies, was obtained by more than 50% of the patients. Decreases in spasms and spasticity, and improved sexual function were also common findings. Clinical improvement seems to be dose-dependent but was not influenced by the chronicity of the SCI. CONCLUSION Personalized cell therapy with MSCs is safe and leads to clear improvements in clinical aspects and quality of life for patients with complete and chronically established paraplegia.
Collapse
Affiliation(s)
- Jesús Vaquero
- Neurosurgery Service, Department of Surgery, Hospital Puerta de Hierro-Majadahonda and Autonomous University, Madrid, Spain; Neurological Cell Therapy Unit, Hospital Puerta de Hierro-Majadahonda and Autonomous University, Madrid, Spain.
| | - Mercedes Zurita
- Neurological Cell Therapy Unit, Hospital Puerta de Hierro-Majadahonda and Autonomous University, Madrid, Spain
| | - Miguel A Rico
- Neurological Cell Therapy Unit, Hospital Puerta de Hierro-Majadahonda and Autonomous University, Madrid, Spain
| | - Celia Bonilla
- Neurological Cell Therapy Unit, Hospital Puerta de Hierro-Majadahonda and Autonomous University, Madrid, Spain
| | - Concepcion Aguayo
- Neurological Cell Therapy Unit, Hospital Puerta de Hierro-Majadahonda and Autonomous University, Madrid, Spain
| | - Jesús Montilla
- Rehabilitation Service, Hospital Puerta de Hierro-Majadahonda and Autonomous University, Madrid, Spain
| | - Salvador Bustamante
- Urology Service, Hospital Puerta de Hierro-Majadahonda and Autonomous University, Madrid, Spain
| | - Joaquin Carballido
- Urology Service, Hospital Puerta de Hierro-Majadahonda and Autonomous University, Madrid, Spain
| | - Esperanza Marin
- Clinical Neurophysiology Service, Hospital Puerta de Hierro-Majadahonda and Autonomous University, Madrid, Spain
| | - Francisco Martinez
- Clinical Neurophysiology Service, Hospital Puerta de Hierro-Majadahonda and Autonomous University, Madrid, Spain
| | - Avelino Parajon
- Neurosurgery Service, Department of Surgery, Hospital Puerta de Hierro-Majadahonda and Autonomous University, Madrid, Spain
| | - Cecilia Fernandez
- Neurosurgery Service, Department of Surgery, Hospital Puerta de Hierro-Majadahonda and Autonomous University, Madrid, Spain
| | - Laura De Reina
- Neurosurgery Service, Department of Surgery, Hospital Puerta de Hierro-Majadahonda and Autonomous University, Madrid, Spain
| | | |
Collapse
|
13
|
Thakkar UG, Vanikar AV, Trivedi HL, Shah VR, Dave SD, Dixit SB, Tiwari BB, Shah HH. Infusion of autologous adipose tissue derived neuronal differentiated mesenchymal stem cells and hematopoietic stem cells in post-traumatic paraplegia offers a viable therapeutic approach. Adv Biomed Res 2016; 5:51. [PMID: 27110548 PMCID: PMC4817398 DOI: 10.4103/2277-9175.178792] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Accepted: 07/11/2015] [Indexed: 12/18/2022] Open
Abstract
Background: Spinal cord injury (SCI) is not likely to recover by current therapeutic modalities. Stem cell (SC) therapy (SCT) has promising results in regenerative medicine. We present our experience of co-infusion of autologous adipose tissue derived mesenchymal SC differentiated neuronal cells (N-Ad-MSC) and hematopoietic SCs (HSCs) in a set of patients with posttraumatic paraplegia. Materials and Methods: Ten patients with posttraumatic paraplegia of mean age 3.42 years were volunteered for SCT. Their mean age was 28 years, and they had variable associated complications. They were subjected to adipose tissue resection for in vitro generation of N-Ad-MSC and bone marrow aspiration for generation of HSC. Generated SCs were infused into the cerebrospinal fluid (CSF) below injury site in all patients. Results: Total mean quantum of SC infused was 4.04 ml with a mean nucleated cell count of 4.5 × 104/μL and mean CD34+ of 0.35%, CD45−/90+ and CD45−/73+ of 41.4%, and 10.04%, respectively. All of them expressed transcription factors beta-3 tubulin and glial fibrillary acid protein. No untoward effect of SCT was noted. Variable and sustained improvement in Hauser's index and American Spinal Injury Association score was noted in all patients over a mean follow-up of 2.95 years. Mean injury duration was 3.42 years against the period of approximately 1-year required for natural recovery, suggesting a positive role of SCs. Conclusion: Co-infusion of N-Ad-MSC and HSC in CSF is safe and viable therapeutic approach for SCIs.
Collapse
Affiliation(s)
- Umang G Thakkar
- Department of Stem Cell Therapy and Regenerative Medicine, G. R. Doshi and K. M. Mehta Institute of Kidney Diseases and Research Centre - Dr. H. L. Trivedi Institute of Transplantation Sciences, Ahmedabad, Gujarat, India
| | - Aruna V Vanikar
- Department of Stem Cell Therapy and Regenerative Medicine, G. R. Doshi and K. M. Mehta Institute of Kidney Diseases and Research Centre - Dr. H. L. Trivedi Institute of Transplantation Sciences, Ahmedabad, Gujarat, India; Department of Pathology, Laboratory Medicine, Transfusion Services and Immunohematology, G. R. Doshi and K. M. Mehta Institute of Kidney Diseases and Research Centre - Dr. H. L. Trivedi Institute of Transplantation Sciences, Ahmedabad, Gujarat, India
| | - Hargovind L Trivedi
- Department of Stem Cell Therapy and Regenerative Medicine, G. R. Doshi and K. M. Mehta Institute of Kidney Diseases and Research Centre - Dr. H. L. Trivedi Institute of Transplantation Sciences, Ahmedabad, Gujarat, India; Department of Nephrology and Transplantation Medicine, G. R. Doshi and K. M. Mehta Institute of Kidney Diseases and Research Centre - Dr. H. L. Trivedi Institute of Transplantation Sciences, Ahmedabad, Gujarat, India
| | - Veena R Shah
- Department of Anesthesiology and Critical Care, G. R. Doshi and K. M. Mehta Institute of Kidney Diseases and Research Centre - Dr. H. L. Trivedi Institute of Transplantation Sciences, Ahmedabad, Gujarat, India
| | - Shruti D Dave
- Department of Pathology, Laboratory Medicine, Transfusion Services and Immunohematology, G. R. Doshi and K. M. Mehta Institute of Kidney Diseases and Research Centre - Dr. H. L. Trivedi Institute of Transplantation Sciences, Ahmedabad, Gujarat, India
| | - Satyajit B Dixit
- Department of Stem Cell Therapy and Regenerative Medicine, G. R. Doshi and K. M. Mehta Institute of Kidney Diseases and Research Centre - Dr. H. L. Trivedi Institute of Transplantation Sciences, Ahmedabad, Gujarat, India
| | - Bharat B Tiwari
- Department of Physiotherapy and Rehabilitation, G. R. Doshi and K. M. Mehta Institute of Kidney Diseases and Research Centre - Dr. H. L. Trivedi Institute of Transplantation Sciences, Ahmedabad, Gujarat, India
| | - Harda H Shah
- Department of Physiotherapy and Rehabilitation, G. R. Doshi and K. M. Mehta Institute of Kidney Diseases and Research Centre - Dr. H. L. Trivedi Institute of Transplantation Sciences, Ahmedabad, Gujarat, India
| |
Collapse
|
14
|
Sandner B, Ciatipis M, Motsch M, Soljanik I, Weidner N, Blesch A. Limited Functional Effects of Subacute Syngeneic Bone Marrow Stromal Cell Transplantation after Rat Spinal Cord Contusion Injury. Cell Transplant 2016; 25:125-39. [DOI: 10.3727/096368915x687679] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Cell transplantation might be one means to improve motor, sensory, or autonomic recovery after traumatic spinal cord injury (SCI). Among the different cell types evaluated to date, bone marrow stromal cells (BMSCs) have received considerable interest due to their potential neuroprotective properties. However, uncertainty exists whether the efficacy of BMSCs after intraspinal transplantation justifies an invasive procedure. In the present study, we analyzed the effect of syngeneic BMSC transplantation following a moderate to severe rat spinal cord injury. Adult Fischer 344 rats underwent a T9 contusion injury (200 kDy) followed by grafting of GFP-expressing BMSCs 3 days postinjury. Animals receiving a contusion injury without cellular grafts or an injury followed by grafts of syngeneic GFP-expressing fibroblasts served as control. Eight weeks post-transplantation, BMSC-grafted animals showed only a minor effect in one measure of sensorimotor recovery, no significant differences in tissue sparing, and no changes in the recovery of bladder function compared to both control groups in urodynamic measurements. Both cell types survived in the lesion site with fibroblasts displaying a larger graft volume. Thus, contrary to some reports using allogeneic or xenogeneic transplants, subacute intraparenchymal grafting of syngeneic BMSCs has only a minor effect on functional recovery.
Collapse
Affiliation(s)
- Beatrice Sandner
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Mareva Ciatipis
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Melanie Motsch
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Irina Soljanik
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Norbert Weidner
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Armin Blesch
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
15
|
Cruz-Martinez P, Pastor D, Estirado A, Pacheco-Torres J, Martinez S, Jones J. Stem cell injection in the hindlimb skeletal muscle enhances neurorepair in mice with spinal cord injury. Regen Med 2015; 9:579-91. [PMID: 25372077 DOI: 10.2217/rme.14.38] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
AIMS To develop a low-risk, little-invasive stem cell-based method to treat acute spinal cord injuries. methods: Adult mice were submitted to an incomplete spinal cord injury, and mesenchymal stem cells injected intramuscularly into both hindlimbs. Behavior tests and MRI of the spinal cord were periodically performed for up to 6 months, along with immunohistochemical analysis. Immunohistochemical and PCR analysis of the muscles were used to detect the grafted cells as well as the soluble factors released. RESULTS The stem cell-treated mice presented significant improvements in their motor skills 5 months after treatment. Spinal cord repair was detected by magnetic resonance and immunohistochemistry. In the hindlimb muscles, the stem cells activated muscle and motor neuron repair mechanisms, due to the secretion of several neurotrophic factors. CONCLUSION Bone marrow mesenchymal stem cell injection into hindlimb muscles stimulates spinal cord repair in acute spinal cord lesions.
Collapse
Affiliation(s)
- Pablo Cruz-Martinez
- Neuroscience Institute, University Miguel Hernández (UMH-CSIC), San Juan, Alicante, Spain
| | | | | | | | | | | |
Collapse
|
16
|
Xiang L, Chen Y. Stem cell transplantation for treating spinal cord injury: A literature comparison between studies of stem cells obtained from various sources. Neural Regen Res 2015; 7:1256-63. [PMID: 25709624 PMCID: PMC4336961 DOI: 10.3969/j.issn.1673-5374.2012.16.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2012] [Accepted: 04/23/2012] [Indexed: 01/09/2023] Open
Abstract
OBJECTIVE: To identify global research trends of stem cell transplantation for treating spinal cord injury using a bibliometric analysis of the Web of Science. DATA RETRIEVAL: We performed a bibliometric analysis of data retrievals for stem cell transplantation for treating spinal cord injury from 2002 to 2011 using the Web of Science. SELECTION CRITERIA: Inclusion criteria: (a) peer-reviewed articles on stem cell transplantation for treating spinal cord injury that were published and indexed in the Web of Science; (b) type of articles: original research articles, reviews, meeting abstracts, proceedings papers, book chapters, editorial material, and news items; and (c) year of publication: 2002–2011. Exclusion criteria: (a) articles that required manual searching or telephone access; (b) documents that were not published in the public domain; and (c) a number of corrected papers from the total number of articles. MAIN OUTCOME MEASURES: (1) Annual publication output; (2) distribution according to country; (3) distribution according to institution; (4) distribution according to journals; (5) distribution according to funding agencies; and (6) top cited articles over the last 10 years. RESULTS: Bone marrow mesenchymal stem cells and embryonic stem cells have been widely used for treating spinal cord injury. In total, 191 studies of bone marrow mesenchymal stem cell transplantation and 236 studies of embryonic stem cell transplantation for treating spinal cord injury appeared in the Web of Science from 2002 to 2011, and almost half of which were derived from American or Japanese authors and institutes. The number of studies of stem cell transplantation for treating spinal cord injury has gradually increased over the past 10 years. Most papers on stem cell transplantation for treating spinal cord injury appeared in journals with a particular focus on stem cell research, such as Stem Cells and Cell Transplantation. Although umbilical cord blood stem cells and adipose-derived stem cells have been studied for treating spinal cord injury, the number of published papers was much smaller, with only 21 and 17 records, respectively, in the Web of Science. CONCLUSION: Based on our analysis of the literature and research trends, we found that stem cells transplantation obtained from various sources have been studied for treating spinal cord injury; however, it is difficult for researchers to reach a consensus on this theme.
Collapse
Affiliation(s)
- Liangbi Xiang
- Department of Orthopedics, General Hospital of Shenyang Military Area Command of Chinese PLA, Shenyang 110016, Liaoning Province, China
| | - Yu Chen
- Department of Orthopedics, General Hospital of Shenyang Military Area Command of Chinese PLA, Shenyang 110016, Liaoning Province, China
| |
Collapse
|
17
|
Li L, Li Y, Jiang H. Neurotrophine-3 may contribute to neuronal differentiation of mesenchymal stem cells through the activation of the bone morphogenetic protein pathway. ACTA ACUST UNITED AC 2015. [DOI: 10.1515/cmble-2015-0023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
AbstractWe investigated whether neurotrophin-3 (NT-3) can promote differentiation of mouse bone mesenchymal stem cells (MSCs) into neurons via the bone morphogenetic protein pathway. MSCs were prepared from rat bone marrow and either transfected with pIRES2-EGFP or pIRES2-EGFP-NT-3 or treated with bone morphogenetic protein 4. The pIRES2-EGFP-NT-3-transfected MSCs further underwent noggin treatment or siRNA-mediated knockout of the TrkC gene or were left untreated. Immunofluorescence staining, real-time PCR and Western blot analyses were performed to evaluate the transcription and expression of neural-specific genes and BMP-Smad signaling. MSCs were efficiently transduced by the NT-3 gene via pIRES2-EGFP vectors. pIRES2- EGFP-NT-3 could initiate the transcription and expression of neural-specific genes, including nestin, NSE and MAP-2, and stimulate BMP-Smad signaling. The transcription and expression of neural-specific genes and BMP-Smad signaling were significantly suppressed by siRNA-mediated knockdown of the TrkC gene of MSCs. These findings suggest that the BMP signaling pathway may be a key regulatory point in NT-3-transfected neuronal differentiation of MSCs. The BMP and neurotrophin pathways contribute to a tightly regulated signaling network that directs the precise connections between neuronal differentiation of MSCs and their targets.
Collapse
|
18
|
Zhang EJ, Song CH, Ko YK, Lee WH. Intrathecal administration of mesenchymal stem cells reduces the reactive oxygen species and pain behavior in neuropathic rats. Korean J Pain 2014; 27:239-45. [PMID: 25031809 PMCID: PMC4099236 DOI: 10.3344/kjp.2014.27.3.239] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 06/13/2014] [Accepted: 06/19/2014] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Neuropathic pain induced by spinal or peripheral nerve injury is very resistant to common pain killers, nerve block, and other pain management approaches. Recently, several studies using stem cells suggested a new way to control the neuropatic pain. In this study, we used the spinal nerve L5 ligation (SNL) model to investigate whether intrathecal rat mesenchymal stem cells (rMSCs) were able to decrease pain behavior, as well as the relationship between rMSCs and reactive oxygen species (ROS). METHODS Neuropathic pain of the left hind paw was induced by unilateral SNL in Sprague-Dawley rats (n = 10 in each group). Mechanical sensitivity was assessed using Von Frey filaments at 3, 7, 10, 12, 14, 17, and 24 days post-ligation. rMSCs (10 µl, 1 × 10(5)) or phosphate buffer saline (PBS, 10 µl) was injected intrathecally at 7 days post-ligation. Dihydroethidium (DHE), an oxidative fluorescent dye, was used to detect ROS at 24 days post-ligation. RESULTS Tight ligation of the L5 spinal nerve induced allodynia in the left hind paw after 3 days post-ligation. ROS expression was increased significantly (P < 0.05) in spinal dorsal horn of L5. Intrathecal rMSCs significantly (P < 0.01) alleviated the allodynia at 10 days after intrathecal injection (17 days post-ligation). Intrathecal rMSCs administration significantly (P < 0.05) reduced ROS expression in the spinal dorsal horn. CONCLUSIONS These results suggest that rMSCs may modulate neuropathic pain generation through ROS expression after spinal nerve ligation.
Collapse
Affiliation(s)
- En Ji Zhang
- Department of Anesthesiology and Pain Medicine, School of Medicine, Chungnam National University, Daejeon, Korea
| | - Chang Hwa Song
- Department of Microbiology, School of Medicine, Chungnam National University, Daejeon, Korea
| | - Young Kwon Ko
- Department of Anesthesiology and Pain Medicine, School of Medicine, Chungnam National University, Daejeon, Korea
| | - Won Hyung Lee
- Department of Anesthesiology and Pain Medicine, School of Medicine, Chungnam National University, Daejeon, Korea
| |
Collapse
|
19
|
Hyatt AJT, Wang D, van Oterendorp C, Fawcett JW, Martin KR. Mesenchymal stromal cells integrate and form longitudinally-aligned layers when delivered to injured spinal cord via a novel fibrin scaffold. Neurosci Lett 2014; 569:12-7. [PMID: 24680849 PMCID: PMC4015360 DOI: 10.1016/j.neulet.2014.03.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2013] [Revised: 03/10/2014] [Accepted: 03/12/2014] [Indexed: 11/11/2022]
Abstract
MSCs can be delivered to injured spinal cord through use of a fibrin scaffold. Scaffold-delivered MSCs form longitudinally-aligned layers over the lesion site. Regenerating axons enter scaffold-delivered grafts and grow longitudinally. MSCs delivered via injection orient perpendicular to the plane of the spinal cord. Regenerating axons in injected grafts grow perpendicular to the plane of the cord.
Mesenchymal stromal cells (MSCs) have been shown to promote healing and regeneration in a number of CNS injury models and therefore there is much interest in the clinical use of these cells. For spinal cord injuries, a standard delivery method for MSCs is intraspinal injection, but this can result in additional injury and provides little control over how the cells integrate into the tissue. The present study examines the use of a novel fibrin scaffold as a new method of delivering MSCs to injured spinal cord. Use of the fibrin scaffold resulted in the formation of longitudinally-aligned layers of MSCs growing over the spinal cord lesion site. Host neurites were able to migrate into this MSC architecture and grow longitudinally. The length of the MSC bridge corresponded to the length of the fibrin scaffold. MSCs that were delivered via intraspinal injection were mainly oriented perpendicular to the plane of the spinal cord and remained largely restricted to the lesion site. Host neurites within the injected MSC graft were also oriented perpendicular to the plane of the spinal cord.
Collapse
Affiliation(s)
- Alex J T Hyatt
- John van Geest Centre for Brain Repair, University of Cambridge, Robinson Way, Cambridge CB2 0PY, United Kingdom
| | - Difei Wang
- John van Geest Centre for Brain Repair, University of Cambridge, Robinson Way, Cambridge CB2 0PY, United Kingdom
| | - Christian van Oterendorp
- John van Geest Centre for Brain Repair, University of Cambridge, Robinson Way, Cambridge CB2 0PY, United Kingdom; University Eye Hospital, Freiburg, Germany
| | - James W Fawcett
- John van Geest Centre for Brain Repair, University of Cambridge, Robinson Way, Cambridge CB2 0PY, United Kingdom
| | - Keith R Martin
- John van Geest Centre for Brain Repair, University of Cambridge, Robinson Way, Cambridge CB2 0PY, United Kingdom; Cambridge NIHR Biomedical Research Centre, UK; Wellcome Trust-MRC Cambridge Stem Cell Institute, UK.
| |
Collapse
|
20
|
The role of mesenchymal stromal cells in spinal cord injury, regenerative medicine and possible clinical applications. Biochimie 2013; 95:2257-70. [DOI: 10.1016/j.biochi.2013.08.004] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2013] [Accepted: 08/05/2013] [Indexed: 12/13/2022]
|
21
|
Oda Y, Tani K, Isozaki A, Haraguchi T, Itamoto K, Nakazawa H, Taura Y. Effects of polyethylene glycol administration and bone marrow stromal cell transplantation therapy in spinal cord injury mice. J Vet Med Sci 2013; 76:415-21. [PMID: 24270802 PMCID: PMC4013369 DOI: 10.1292/jvms.13-0167] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Bone marrow stromal cell (BMSC) transplantation has been reported as treatments
that promote functional recovery after spinal cord injury (SCI) in humans and animals.
Polyethylene glycol (PEG) has been also reported as treatments that promote functional
recovery after spinal cord injury (SCI) in humans and animals. Therefore, administration
of PEG combined with BMSC transplantation may improve outcomes compared with BMSC
transplantation only in SCI model mice. SCI mice were divided into a control-group,
BMSC-group, PEG-group and BMSC+PEG-group. BMSC transplantation and PEG administration were
performed immediately after surgery. Compared to the control-group, PEG- and
BMSC+PEG-groups showed significant locomotor functional recovery 4 weeks after therapy. We
observed no significant differences among the groups. In the BMSC- and BMSC+PEG-groups,
immunohistochemistry showed that many neuronal cells aggressively migrated toward the
glial scar from the region rostral of the lesion site. In the control- and PEG-groups, the
boundary of the injured regions was covered with astrocytes, and a few neuronal cells were
migrated toward the glial scar. We conclude that combined BMSC transplantation with PEG
treatment showed no synergistic effects on locomotor functional recovery or beneficial
cellular events. Further studies may improve the effect of the treatment, including
modification of the timing of BMSC transplantation.
Collapse
Affiliation(s)
- Yasutaka Oda
- Department of Veterinary Surgery, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1 Yoshida, Yamaguchi 753-8515, Japan
| | | | | | | | | | | | | |
Collapse
|
22
|
Vaquero J, Zurita M. Cell transplantation in paraplegic patients: the importance of properly assessing the spinal cord morphology. Clin Transplant 2013; 27:968-71. [DOI: 10.1111/ctr.12267] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/19/2013] [Indexed: 11/30/2022]
Affiliation(s)
- Jesús Vaquero
- Neuroscience Research Unit of Neurosurgical Department; Hospital Puerta de Hierro-Majadahonda; Rafael del Pino Chair and UAM-Los Madroños Hospital Chair; Autonomous University; Madrid Spain
| | - Mercedes Zurita
- Neuroscience Research Unit of Neurosurgical Department; Hospital Puerta de Hierro-Majadahonda; Rafael del Pino Chair and UAM-Los Madroños Hospital Chair; Autonomous University; Madrid Spain
| |
Collapse
|
23
|
Zurita M, Aguayo C, Bonilla C, Rodriguez A, Vaquero J. Perilesional intrathecal administration of autologous bone marrow stromal cells achieves functional improvement in pigs with chronic paraplegia. Cytotherapy 2013; 15:1218-27. [DOI: 10.1016/j.jcyt.2013.05.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Revised: 04/11/2013] [Accepted: 05/16/2013] [Indexed: 10/26/2022]
|
24
|
Vaquero J, Otero L, Bonilla C, Aguayo C, Rico MA, Rodriguez A, Zurita M. Cell therapy with bone marrow stromal cells after intracerebral hemorrhage: impact of platelet-rich plasma scaffolds. Cytotherapy 2013; 15:33-43. [PMID: 23260084 DOI: 10.1016/j.jcyt.2012.10.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Accepted: 07/19/2012] [Indexed: 01/27/2023]
Abstract
BACKGROUND AIMS Cell therapy using bone marrow stromal cells (BMSCs) has been considered a promising strategy for neurologic sequelae after intracerebral hemorrhage (ICH). However, after intracerebral administration of BMSCs, most of the cells die, partly because of the absence of extracellular matrix. Intracerebral transplantation of BMSCs, supported in a platelet-rich plasma (PRP) scaffold, optimizes this type of cell therapy. METHODS ICH was induced by stereotactic injection of 0.5 IU of collagenase type IV in the striatum of adult Wistar rats (n = 40). Two months later, the rats were subjected to intracerebral administration of 5 × 10(6) allogeneic BMSCs embedded in a PRP scaffold (n = 10), 5 × 10(6) allogeneic BMSCs in saline (n = 10), PRP-derived scaffold only (n = 10) or saline only (n = 10). Functional improvements in each group over the next 6 months were assessed using Rotarod and Video-Tracking-Box tests. Endogenous neurogenesis and survival of transplanted BMSCs were examined at the end of follow-up. RESULTS Our study demonstrated neurologic improvement after BMSC transplantation and significantly better functional improvement for the group of animals that received BMSCs in the PRP-derived scaffold compared with the group that received BMSCs in saline. Histologic results showed that better functional outcome was associated with strong activation of endogenous neurogenesis. After intracerebral administration of BMSCs, donor cells were integrated in the injured tissue and showed phenotypic expression of glial fibrillary acidic protein and neuronal nucleus. CONCLUSIONS PRP-derived scaffolds increase the viability and biologic activity of BMSCs and optimize functional recovery when this type of cell therapy is applied after ICH.
Collapse
Affiliation(s)
- Jesus Vaquero
- Neuroscience Research Unit, Neuroscience Rafael del Pino Chair, and Neurosurgical Service, Hospital Puerta de Hierro-Majadahonda, Autonomous University, Madrid, Spain.
| | | | | | | | | | | | | |
Collapse
|
25
|
Hejčl A, Růžička J, Kapcalová M, Turnovcová K, Krumbholcová E, Přádný M, Michálek J, Cihlář J, Jendelová P, Syková E. Adjusting the chemical and physical properties of hydrogels leads to improved stem cell survival and tissue ingrowth in spinal cord injury reconstruction: a comparative study of four methacrylate hydrogels. Stem Cells Dev 2013; 22:2794-805. [PMID: 23750454 DOI: 10.1089/scd.2012.0616] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Currently, there is no effective strategy for the treatment of spinal cord injury (SCI). A suitable combination of modern hydrogel materials, modified to effectively bridge the lesion cavity, combined with appropriate stem cell therapy seems to be a promising approach to repair spinal cord damage. We demonstrate the synergic effect of porosity and surface modification of hydrogels on mesenchymal stem cell (MSC) adhesiveness in vitro and their in vivo survival in an experimental model of SCI. MSCs were seeded on four different hydrogels: hydroxypropylmethacrylate-RGD prepared by heterophase separation (HPMA-HS-RGD) and three other hydrogels polymerized in the presence of a solid porogen: HPMA-SP, HPMA-SP-RGD, and hydroxy ethyl methacrylate [2-(methacryloyloxy)ethyl] trimethylammonium chloride (HEMA-MOETACl). Their adhesion capability and cell survival were evaluated at 1, 7, and 14 days after the seeding of MSCs on the hydrogel scaffolds. The cell-polymer scaffolds were then implanted into hemisected rat spinal cord, and MSC survival in vivo and the ingrowth of endogenous tissue elements were evaluated 1 month after implantation. In vitro data demonstrated that HEMA-MOETACl and HPMA-SP-RGD hydrogels were superior in the number of cells attached. In vivo, the highest cell survival was found in the HEMA-MOETACl hydrogels; however, only a small ingrowth of blood vessels and axons was observed. Both HPMA-SP and HPMA-SP-RGD hydrogels showed better survival of MSCs compared with the HPMA-HS-RGD hydrogel. The RGD sequence attached to both types of HPMA hydrogels significantly influenced the number of blood vessels inside the implanted hydrogels. Further, the porous structure of HPMA-SP hydrogels promoted a statistically significant greater ingrowth of axons and less connective tissue elements into the implant. Our results demonstrate that the physical and chemical properties of the HPMA-SP-RGD hydrogel show the best combination for bridging a spinal cord lesion, while the HEMA-MOETACl hydrogel serves as the best carrier of MSCs.
Collapse
Affiliation(s)
- Aleš Hejčl
- 1 Department of Neuroscience, Institute of Experimental Medicine , Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
A comparison of the behavioral and anatomical outcomes in sub-acute and chronic spinal cord injury models following treatment with human mesenchymal precursor cell transplantation and recombinant decorin. Exp Neurol 2013; 248:343-59. [PMID: 23867131 DOI: 10.1016/j.expneurol.2013.06.018] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Revised: 06/17/2013] [Accepted: 06/19/2013] [Indexed: 01/01/2023]
Abstract
This study assessed the potential of highly purified (Stro-1(+)) human mesenchymal precursor cells (hMPCs) in combination with the anti-scarring protein decorin to repair the injured spinal cord (SC). Donor hMPCs isolated from spinal cord injury (SCI) patients were transplanted into athymic rats as a suspension graft, alone or after previous treatment with, core (decorin(core)) and proteoglycan (decorin(pro)) isoforms of purified human recombinant decorin. Decorin was delivered via mini-osmotic pumps for 14 days following sub-acute (7 day) or chronic (1 month) SCI. hMPCs were delivered to the spinal cord at 3 weeks or 6 weeks after the initial injury at T9 level. Behavioral and anatomical analysis in this study showed statistically significant improvement in functional recovery, tissue sparing and cyst volume reduction following hMPC therapy. The combination of decorin infusion followed by hMPC therapy did not improve these measured outcomes over the use of cell therapy alone, in either sub-acute or chronic SCI regimes. However, decorin infusion did improve tissue sparing, reduce spinal tissue cavitation and increase transplanted cell survivability as compared to controls. Immunohistochemical analysis of spinal cord sections revealed differences in glial, neuronal and extracellular matrix molecule expression within each experimental group. hMPC transplanted spinal cords showed the increased presence of serotonergic (5-HT) and sensory (CGRP) axonal growth within and surrounding transplanted hMPCs for up to 2 months; however, no evidence of hMPC transdifferentiation into neuronal or glial phenotypes. The number of hMPCs was dramatically reduced overall, and no transplanted cells were detected at 8 weeks post-injection using lentiviral GFP labeling and human nuclear antigen antibody labeling. The presence of recombinant decorin in the cell transplantation regimes delayed in part the loss of donor cells, with small numbers remaining at 2 months after transplantation. In vitro co-culture experiments with embryonic dorsal root ganglion explants revealed the growth promoting properties of hMPCs. Decorin did not increase axonal outgrowth from that achieved by hMPCs. We provide evidence for the first time that (Stro-1(+)) hMPCs provide: i) an advantageous source of allografts for stem cell transplantation for sub-acute and chronic spinal cord therapy, and (ii) a positive host microenvironment that promotes tissue sparing/repair that subsequently improves behavioral outcomes after SCI. This was not measurably improved by recombinant decorin treatment, but does provide important information for the future development and potential use of decorin in contusive SCI therapy.
Collapse
|
27
|
Barriga A, Medrano M, De-Juan J, Burgos J. [Intravenous infusion of adult adipose tissue stem cells for repairing spinal cord ischaemic lesions. An experimental study on animals]. Rev Esp Cir Ortop Traumatol (Engl Ed) 2013; 57:89-94. [PMID: 23608207 DOI: 10.1016/j.recot.2013.01.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2013] [Revised: 01/16/2013] [Accepted: 01/17/2013] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE To assess if a peripheral intravenous infusion of adipose tissue stem cells (ATSC), after an ischemic spinal cord injury can promote selective cell migration and cell survival in the damaged neural tissue. ANIMALS AND METHOD: An ischaemic spinal cord injury was provoked by trapping the abdominal aorta for 20 minutes in 11 male New Zealand rabbits (2.5±0.5kg). They were randomised into two groups: one group (n=5) received an intravenous transfusion of 10±2×10(6) ATSC at 24 hours from the injury, and the control group (n=6) were only given the vehicle. The functional status was assessed, using the Tarlov scale at 24h, and 7 and 14 days. The animals were sacrificed at 14 days and a histological and immunohistochemical study was performed. RESULTS Complete paraplegia was achieved in both groups. There were no significant differences as regards neurological recovery, which was nil in both cases. In the histological and immunohistochemical study, it was tested to see if there was any bromodeoxyuridine-marked ATSC in the area of the lesion, but there was only a small amount. CONCLUSION ATSC are able to migrate and survive in the injured spinal cord after aortic ischaemia after they have been administered intravenously. Intravenous infusion is a harmless procedure with no side effect. No neurological recovery was achieved.
Collapse
Affiliation(s)
- A Barriga
- Servicio de Cirugía Ortopédica y Traumatología, Hospital Nacional de Parapléjicos, Toledo, España; Universidad de Castilla-La Mancha, Toledo, España.
| | | | | | | |
Collapse
|
28
|
Intravenous infusion of adult adipose tissue stem cells for repairing spinal cord ischaemic lesions. An experimental study on animals. Rev Esp Cir Ortop Traumatol (Engl Ed) 2013. [DOI: 10.1016/j.recote.2013.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
29
|
Falavigna A, da Costa JC. Mesenchymal autologous stem cells. World Neurosurg 2013; 83:236-50. [PMID: 23402865 DOI: 10.1016/j.wneu.2013.02.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Revised: 01/24/2013] [Accepted: 02/05/2013] [Indexed: 02/07/2023]
Abstract
The use of cell-based therapies for spinal cord injuries has recently gained prominence as a potential therapy or component of a combination strategy. Experimental and clinical studies have been performed using mesenchymal stem cell therapy to treat spinal cord injuries with encouraging results. However, there have been reports on the adverse effects of these stem cell-based therapies, especially in the context of tumor modulation. This article surveys the literature relevant to the potential of mesenchymal autologous stem cells for spinal cord injuries and their clinical implications.
Collapse
Affiliation(s)
- Asdrubal Falavigna
- Department of Neurosurgery, Medical School of the University of Caxias do Sul, Caxias do Sul, Brazil.
| | - Jaderson Costa da Costa
- Neurology Service and Instituto do Cérebro, Pontifical Catholic University of Rio Grande do Sul, Brazil
| |
Collapse
|
30
|
Naghdi M, Tiraihi T, Mesbah-Namin SA, Arabkharadmand J, Kazemi H, Taheri T. Improvement of Contused Spinal Cord in Rats by Cholinergic-like Neuron Therapy. IRANIAN RED CRESCENT MEDICAL JOURNAL 2013; 15:127-35. [PMID: 23682324 PMCID: PMC3652499 DOI: 10.5812/ircmj.7653] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Accepted: 01/08/2013] [Indexed: 12/28/2022]
Abstract
Background Disability in spinal cord injury is an important medical problem, and cell transplantation is considered as an option for the treatment. Objectives The purpose of this study is to use bone marrow stromal cells (BMSCs) derived cholinergic neuron-like cells (CNL) in order to ameliorate the contusion model of spinal cord injury in rats. Materials and Methods The CNLs were produced by pre inducing BMSCs with β-mercaptoethanol (BME) followed by inducing with nerve growth factor (NGF). The cells were immunoreactive to neurofilament 200, NeuN, synaptophysin, synapsin, microtubule associated protein-2 and choline acetyl transferase (ChAT). The CNL were transplanted in contused rats (CR), which were sacrificed after 12 weeks. Results The results showed that BBB test showed an improvement in the CR, while the quantitative analysis showed that the improvement rate was higher in the rats treated with CNL than those treated with BMSCs only or the untreated animals, similar results were noticed in the improvement index. Immunohistochemical analysis of the tissue section prepared from the CR showed that the transplanted cells were engrafted and integrated in the traumatized spinal cord. The morphometric analysis showed that the volume density of the cavity in the CNL treated rats was significantly lower than that of the untreated ones, while the spinal tissue regeneration index was significantly higher. Conclusions The conclusion of the study is that CNL can improve the injured spinal cord.
Collapse
Affiliation(s)
- Majid Naghdi
- Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, IR Iran
| | - Taki Tiraihi
- Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University; Shefa Neuroscience Research Center, Khatam Al-anbia Hospital, Tehran, IR Iran
- Corresponding author: Taki Tiraihi, Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, P.O: 14155-4838, Tehran, IR Iran. Tel: +98-2183553920, Fax: +98-2183553920, E-mail:
| | - Seyed Alireza Mesbah-Namin
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, IR Iran
| | | | - Hadi Kazemi
- Shefa Neuroscience Research Center, Khatam Al-anbia Hospital, Tehran, IR Iran
| | - Taher Taheri
- Shefa Neuroscience Research Center, Khatam Al-anbia Hospital, Tehran, IR Iran
| |
Collapse
|
31
|
Choi JS, Leem JW, Lee KH, Kim SS, Suh-Kim H, Jung SJ, Kim UJ, Lee BH. Effects of human mesenchymal stem cell transplantation combined with polymer on functional recovery following spinal cord hemisection in rats. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2012; 16:405-11. [PMID: 23269903 PMCID: PMC3526745 DOI: 10.4196/kjpp.2012.16.6.405] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Revised: 10/12/2012] [Accepted: 10/22/2012] [Indexed: 12/23/2022]
Abstract
The spontaneous axon regeneration of damaged neurons is limited after spinal cord injury (SCI). Recently, mesenchymal stem cell (MSC) transplantation was proposed as a potential approach for enhancing nerve regeneration that avoids the ethical issues associated with embryonic stem cell transplantation. As SCI is a complex pathological entity, the treatment of SCI requires a multipronged approach. The purpose of the present study was to investigate the functional recovery and therapeutic potential of human MSCs (hMSCs) and polymer in a spinal cord hemisection injury model. Rats were subjected to hemisection injuries and then divided into three groups. Two groups of rats underwent partial thoracic hemisection injury followed by implantation of either polymer only or polymer with hMSCs. Another hemisection-only group was used as a control. Behavioral, electrophysiological and immunohistochemical studies were performed on all rats. The functional recovery was significantly improved in the polymer with hMSC-transplanted group as compared with control at five weeks after transplantation. The results of electrophysiologic study demonstrated that the latency of somatosensory-evoked potentials (SSEPs) in the polymer with hMSC-transplanted group was significantly shorter than in the hemisection-only control group. In the results of immunohistochemical study, β-gal-positive cells were observed in the injured and adjacent sites after hMSC transplantation. Surviving hMSCs differentiated into various cell types such as neurons, astrocytes and oligodendrocytes. These data suggest that hMSC transplantation with polymer may play an important role in functional recovery and axonal regeneration after SCI, and may be a potential therapeutic strategy for SCI.
Collapse
Affiliation(s)
- Ji Soo Choi
- Department of Physiology, Brain Korea 21 Project for Medical Science, and Brain Research Institute, Yonsei University College of Medicine, Seoul 120-752, Korea
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Nery AA, Nascimento IC, Glaser T, Bassaneze V, Krieger JE, Ulrich H. Human mesenchymal stem cells: from immunophenotyping by flow cytometry to clinical applications. Cytometry A 2012; 83:48-61. [PMID: 23027703 DOI: 10.1002/cyto.a.22205] [Citation(s) in RCA: 96] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Revised: 08/15/2012] [Accepted: 08/22/2012] [Indexed: 12/25/2022]
Abstract
Modern medicine will unequivocally include regenerative medicine as a major breakthrough in the re-establishment of damaged or lost tissues due to degenerative diseases or injury. In this scenario, millions of patients worldwide can have their quality of life improved by stem cell implantation coupled with endogenous secretion or administration of survival and differentiation promoting factors. Large efforts, relying mostly on flow cytometry and imaging techniques, have been put into cell isolation, immunophenotyping, and studies of differentiation properties of stem cells of diverse origins. Mesenchymal stem cells (MSCs) are particularly relevant for therapy due to their simplicity of isolation. A minimal phenotypic pattern for the identification of MSCs cells requires them to be immunopositive for CD73, CD90, and CD105 expression, while being negative for CD34, CD45, and HLA-DR and other surface markers. MSCs identified by their cell surface marker expression pattern can be readily purified from patient's bone marrow and adipose tissues. Following expansion and/or predifferentiation into a desired tissue type, stem cells can be reimplanted for tissue repair in the same patient, virtually eliminating rejection problems. Transplantation of MSCs is subject of almost 200 clinical trials to cure and treat a very broad range of conditions, including bone, heart, and neurodegenerative diseases. Immediate or medium term improvements of clinical symptoms have been reported as results of many clinical studies.
Collapse
Affiliation(s)
- Arthur A Nery
- Departamento de Bioquímica, Universidade de São Paulo, São Paulo, Brazil
| | | | | | | | | | | |
Collapse
|
33
|
Patel AN, Vargas V, Revello P, Bull DA. Mesenchymal stem cell population isolated from the subepithelial layer of umbilical cord tissue. Cell Transplant 2012; 22:513-9. [PMID: 23057960 DOI: 10.3727/096368912x655064] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The therapeutic use of stem cells to treat diseases and injuries is a promising tool in regenerative medicine. The umbilical cord provides a rich source of stem cells; we have previously reported a population of stem cells isolated from Wharton's jelly. In this report, we aimed to isolate a novel cell population that was different than those found in Wharton's jelly. We isolated stem cells from the subepithelial layer of the umbilical cord; the cells could be expanded for greater than 90 population doubling and had mesenchymal stem cell characteristics, expressing CD9, SSEA4, CD44, CD90, CD166, CD73, and CD146 but were negative for STRO-1. The cells can be directionally differentiated and undergo osteo-, chondro-, adipo-, and cardiogenesis. In addition, we have identified for the first time that mesenchymal stem cells isolated from umbilical cord can produce microvesicles, termed exosomes. This is the first report describing a stem cell population isolated from the subepithelial layer of the umbilical cord. Given the growth capacity, multilineage potential, and most importantly the low levels of HLA-ABC, we propose that this novel cell isolated from the subepithelial layer of umbilical cord is an ideal candidate for allogeneic cell-based therapy.
Collapse
Affiliation(s)
- Amit N Patel
- Division of Cardiothoracic Surgery, University of Utah, Salt Lake City, UT, USA.
| | | | | | | |
Collapse
|
34
|
Park JH, Kim DY, Sung IY, Choi GH, Jeon MH, Kim KK, Jeon SR. Long-term results of spinal cord injury therapy using mesenchymal stem cells derived from bone marrow in humans. Neurosurgery 2012; 70:1238-47; discussion 1247. [PMID: 22127044 DOI: 10.1227/neu.0b013e31824387f9] [Citation(s) in RCA: 132] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Although the transplantation of mesenchymal stem cells (MSCs) after spinal cord injury (SCI) has shown promising results in animals, less is known about the effects of autologous MSCs in human SCI. OBJECTIVE To describe the long-term results of 10 patients who underwent intramedullary direct MSCs transplantation into injured spinal cords. METHODS Autologous MSCs were harvested from the iliac bone of each patient and expanded by culturing for 4 weeks. MSCs (8 × 10) were directly injected into the spinal cord, and 4 × 10 cells were injected into the intradural space of 10 patients with American Spinal Injury Association class A or B injury caused by traumatic cervical SCI. After 4 and 8 weeks, an additional 5 × 10 MSCs were injected into each patient through lumbar tapping. Outcome assessments included changes in the motor power grade of the extremities, magnetic resonance imaging, and electrophysiological recordings. RESULTS Although 6 of the 10 patients showed motor power improvement of the upper extremities at 6-month follow-up, 3 showed gradual improvement in activities of daily living, and changes on magnetic resonance imaging such as decreases in cavity size and the appearance of fiber-like low signal intensity streaks. They also showed electrophysiological improvement. All 10 patients did not experience any permanent complication associated with MSC transplantation. CONCLUSION Three of the 10 patients with SCI who were directly injected with autologous MSCs showed improvement in the motor power of the upper extremities and in activities of daily living, as well as significant magnetic resonance imaging and electrophysiological changes during long-term follow-up.
Collapse
Affiliation(s)
- Jin Hoon Park
- Department of Neurological Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | | | | | | | | | | | | |
Collapse
|
35
|
It takes two to tango: activation of cortex and lumbosacral circuitry restores locomotion in spinal cord injury. World Neurosurg 2012; 78:380-3. [PMID: 22960539 DOI: 10.1016/j.wneu.2012.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
36
|
Otero L, Zurita M, Bonilla C, Aguayo C, Rico MA, Vaquero J. [Perspectives of cell therapy in sequelae from cerebrovascular accidents]. Neurocirugia (Astur) 2012; 23:193-9. [PMID: 22858052 DOI: 10.1016/j.neucir.2011.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Accepted: 10/28/2011] [Indexed: 11/15/2022]
Abstract
INTRODUCTION Spontaneous intracerebral hemorrhage (ICH) is associated with mortality between 40 and 50% of cases. Among the survivors, only 10% are independent after one month, there is no effective treatment of sequelae, except for the limited possibilities providing for rehabilitation. OBJECTIVES We review the current experience with intracerebral transplantation of mesenchymal stem cells (MSCs) obtained from bone marrow as a potential treatment of neurological sequelae occurring after experimental ICH. MATERIAL AND METHODS We describe the model of ICH by intracerebral administration of collagenaseIV at basal ganglia level in Wistar rats. Neurological deficits caused by ICH can be quantified through a variety of functional assessment test (NMSS, Rota-rod, VTB-test). 5×10allogeneic MSCs in 10μl of saline were administered intracerebrally in 10 animals, 2 months after ICH. In another 10 animals (controls) the same volume of saline was administered. Changes in the functional deficits were assessed during the next 6 months in both experimental groups. RESULTS The results suggested therapeutic efficacy of MSCs transplantation and showed that transplanted stem cells can survive in the injured brain, transforming into neurons and glial cells. This form of cell therapy induces reactivation of endogenous neurogenesis at the subventricular zone (SVZ) and achieves antiapoptotic protective effect in the injured brain. CONCLUSIONS Cell therapy represents an important field of research with potential clinical application to treatment of neurological sequels, currently considered irreversible. Neurosurgeons should become involved in the development of these new techniques that are likely to shape the future of this specialty.
Collapse
Affiliation(s)
- Laura Otero
- Unidad de Investigación en Neurociencias y Cátedra Rafael del Pino de Neurociencias, Servicio de Neurocirugía, Hospital Universitario Puerta de Hierro, Majadahonda, Madrid, España
| | | | | | | | | | | |
Collapse
|
37
|
Ritfeld GJ, Tewarie RDSN, Vajn K, Rahiem ST, Hurtado A, Wendell DF, Roos RAC, Oudega M. Bone Marrow Stromal Cell-Mediated Tissue Sparing Enhances Functional Repair after Spinal Cord Contusion in Adult Rats. Cell Transplant 2012; 21:1561-75. [DOI: 10.3727/096368912x640484] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Bone marrow stromal cell (BMSC) transplantation has shown promise for repair of the spinal cord. We showed earlier that a BMSC transplant limits the loss of spinal nervous tissue after a contusive injury. Here, we addressed the premise that BMSC-mediated tissue sparing underlies functional recovery in adult rats after a contusion of the thoracic spinal cord. Our results reveal that after 2 months BMSCs had elicited a significant increase in spared tissue volumes and in blood vessel density in the contusion epicenter. A strong functional relationship existed between spared tissue volumes and blood vessel density. BMSC-transplanted rats exhibited significant improvements in motor, sensorimotor, and sensory functions, which were strongly correlated with spared tissue volumes. Retrograde tracing revealed that rats with BMSCs had twice as many descending brainstem neurons with an axon projecting beyond the contused spinal cord segment and these correlated strongly with the improved motor/sensorimotor functions but not sensory functions. Together, our data indicate that tissue sparing greatly contributes to BMSC-mediated functional repair after spinal cord contusion. The preservation/formation of blood vessels and sparing/regeneration of descending brainstem axons may be important mediators of the BMSC-mediated anatomical and functional improvements.
Collapse
Affiliation(s)
- Gaby J. Ritfeld
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Katarina Vajn
- Department of Neurosurgery, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
| | - Sahar T. Rahiem
- International Center for Spinal Cord Injury, Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD, USA
| | - Andres Hurtado
- International Center for Spinal Cord Injury, Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dane F. Wendell
- International Center for Spinal Cord Injury, Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD, USA
| | - Raymund A. C. Roos
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Martin Oudega
- Department of Physical Medicine & Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Bioengineering, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
38
|
Quertainmont R, Cantinieaux D, Botman O, Sid S, Schoenen J, Franzen R. Mesenchymal stem cell graft improves recovery after spinal cord injury in adult rats through neurotrophic and pro-angiogenic actions. PLoS One 2012; 7:e39500. [PMID: 22745769 PMCID: PMC3380009 DOI: 10.1371/journal.pone.0039500] [Citation(s) in RCA: 153] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Accepted: 05/21/2012] [Indexed: 12/13/2022] Open
Abstract
Numerous strategies have been managed to improve functional recovery after spinal cord injury (SCI) but an optimal strategy doesn't exist yet. Actually, it is the complexity of the injured spinal cord pathophysiology that begets the multifactorial approaches assessed to favour tissue protection, axonal regrowth and functional recovery. In this context, it appears that mesenchymal stem cells (MSCs) could take an interesting part. The aim of this study is to graft MSCs after a spinal cord compression injury in adult rat to assess their effect on functional recovery and to highlight their mechanisms of action. We found that in intravenously grafted animals, MSCs induce, as early as 1 week after the graft, an improvement of their open field and grid navigation scores compared to control animals. At the histological analysis of their dissected spinal cord, no MSCs were found within the host despite their BrdU labelling performed before the graft, whatever the delay observed: 7, 14 or 21 days. However, a cytokine array performed on spinal cord extracts 3 days after MSC graft reveals a significant increase of NGF expression in the injured tissue. Also, a significant tissue sparing effect of MSC graft was observed. Finally, we also show that MSCs promote vascularisation, as the density of blood vessels within the lesioned area was higher in grafted rats. In conclusion, we bring here some new evidences that MSCs most likely act throughout their secretions and not via their own integration/differentiation within the host tissue.
Collapse
Affiliation(s)
- Renaud Quertainmont
- GIGA Neurosciences, Axonal Regeneration and Cephalic Pain Unit, University of Liege, Liege, Belgium
| | - Dorothée Cantinieaux
- GIGA Neurosciences, Axonal Regeneration and Cephalic Pain Unit, University of Liege, Liege, Belgium
| | - Olivier Botman
- GIGA Neurosciences, Axonal Regeneration and Cephalic Pain Unit, University of Liege, Liege, Belgium
| | - Selim Sid
- GIGA Neurosciences, Axonal Regeneration and Cephalic Pain Unit, University of Liege, Liege, Belgium
| | - Jean Schoenen
- GIGA Neurosciences, Axonal Regeneration and Cephalic Pain Unit, University of Liege, Liege, Belgium
| | - Rachelle Franzen
- GIGA Neurosciences, Axonal Regeneration and Cephalic Pain Unit, University of Liege, Liege, Belgium
- * E-mail:
| |
Collapse
|
39
|
The pig model of chronic paraplegia: A challenge for experimental studies in spinal cord injury. Prog Neurobiol 2012; 97:288-303. [DOI: 10.1016/j.pneurobio.2012.04.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Revised: 01/22/2012] [Accepted: 04/17/2012] [Indexed: 12/27/2022]
|
40
|
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: 3.5] [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
|
41
|
Nishida H, Nakayama M, Tanaka H, Kitamura M, Hatoya S, Sugiura K, Harada Y, Suzuki Y, Ide C, Inaba T. Safety of autologous bone marrow stromal cell transplantation in dogs with acute spinal cord injury. Vet Surg 2012; 41:437-42. [PMID: 22548465 DOI: 10.1111/j.1532-950x.2011.00959.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
OBJECTIVE To assess the feasibility and safety of transplantation of autologous bone marrow stromal cell (BMSC) in dogs with acute spinal cord injury (SCI). STUDY DESIGN An open-label single-arm trial. ANIMALS Dogs (n = 7) with severe SCI from T6 to L5, caused by vertebral fracture and luxation. METHODS Decompressive and stabilization surgery was performed on dogs with severe SCI caused by vertebral fracture and luxation. Autologous BMSCs were obtained from each dog's femur, cultured, and then injected into the lesion in the acute stage. Adverse events and motor and sensory function were observed for >1 year after SCI. RESULTS Follow-up was 29-62 months after SCI. No complications (eg, infection, neuropathic pain, worsening of neurologic function) were observed. Two dogs walked without support, but none of the 7 dogs had any change in sensory function. CONCLUSIONS Autologous BMSC transplantation is feasible and safe in dogs with acute SCI. Further studies are needed to determine the efficacy of this therapy.
Collapse
|
42
|
He J, Wang XM, Spector M, Cui FZ. Scaffolds for central nervous system tissue engineering. FRONTIERS OF MATERIALS SCIENCE 2012; 6:1-25. [DOI: 10.1007/s11706-012-0157-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
43
|
Bonilla C, Zurita M, Otero L, Aguayo C, Rico MA, Rodríguez A, Vaquero J. Failure of Delayed Intravenous Administration of Bone Marrow Stromal Cells after Traumatic Brain Injury. J Neurotrauma 2012; 29:394-400. [DOI: 10.1089/neu.2011.2101] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Affiliation(s)
- Celia Bonilla
- Neuroscience Research Unit, Puerta de Hierro-Majadahonda Hospital, Madrid, Spain
| | - Mercedes Zurita
- Neuroscience Research Unit, Puerta de Hierro-Majadahonda Hospital, Madrid, Spain
| | - Laura Otero
- Neuroscience Research Unit, Puerta de Hierro-Majadahonda Hospital, Madrid, Spain
| | - Concepción Aguayo
- Neuroscience Research Unit, Puerta de Hierro-Majadahonda Hospital, Madrid, Spain
| | - Miguel A. Rico
- Neuroscience Research Unit, Puerta de Hierro-Majadahonda Hospital, Madrid, Spain
| | - Alicia Rodríguez
- Neuroscience Research Unit, Puerta de Hierro-Majadahonda Hospital, Madrid, Spain
| | - Jesús Vaquero
- Neuroscience Research Unit, Puerta de Hierro-Majadahonda Hospital, Madrid, Spain
- Service of Neurosurgery, Puerta de Hierro-Majadahonda Hospital, Madrid, Spain
| |
Collapse
|
44
|
Affiliation(s)
- R Vawda
- Department of University Health Network, Toronto Western Hospital, Toronto, Canada, ON M5T 2S8
| | - J Wilcox
- Department of University Health Network, Toronto Western Hospital, Toronto, Canada, ON M5T 2S8
| | - MG Fehlings
- Department of University Health Network, Toronto Western Hospital, Toronto, Canada, ON M5T 2S8
| |
Collapse
|
45
|
Wright KT, El Masri W, Osman A, Chowdhury J, Johnson WEB, Franchina M, Lanzoni G, Cantoni S, Cavallini C, Bianchi F, Tazzari PL, Pasquinelli G, Foroni L, Ventura C, Grossi A, Bagnara GP. Concise review: Bone marrow for the treatment of spinal cord injury: mechanisms and clinical applications. Stem Cells 2011; 29:169-78. [PMID: 21732476 PMCID: PMC3083520 DOI: 10.1002/stem.570] [Citation(s) in RCA: 164] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Transplantation of bone marrow stem cells into spinal cord lesions enhances axonal regeneration and promotes functional recovery in animal studies. There are two types of adult bone marrow stem cell; hematopoietic stem cells (HSCs), and mesenchymal stem cells (MSCs). The mechanisms by which HSCs and MSCs might promote spinal cord repair following transplantation have been extensively investigated. The objective of this review is to discuss these mechanisms; we briefly consider the controversial topic of HSC and MSC transdifferentiation into central nervous system cells but focus on the neurotrophic, tissue sparing, and reparative action of MSC grafts in the context of the spinal cord injury (SCI) milieu. We then discuss some of the specific issues related to the translation of HSC and MSC therapies for patients with SCI and present a comprehensive critique of the current bone marrow cell clinical trials for the treatment of SCI to date.
Collapse
Affiliation(s)
- Karina T Wright
- Spinal Studies and Midlands Centre for Spinal Injuries, RJAH Orthopaedic Hospital, Oswestry, Shropshire, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Intravenously transplanted bone marrow stromal cells promote recovery of lower urinary tract function in rats with complete spinal cord injury. Spinal Cord 2011; 50:202-7. [PMID: 22124344 DOI: 10.1038/sc.2011.128] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
OBJECTIVES There is increasing evidence that intravenously injected neural progenitor cells promote recovery of bladder function in rodents, following contusive spinal cord injury through migrating into the injured spinal cord tissue and differentiating into central nervous system cells. The present study was aimed to clarify whether intravenously transplanted bone marrow stromal cells (BMSCs) could improve lower urinary tract (LUT) function in rats with spinal cord transection (SCT). METHODS A total of 22 rats underwent experimentation in three groups, including group 1-sham operation, group 2 (BMSC)-SCT plus BrdU (5-bromo-2'-deoxyuridine) labeled BMSCs transplantation at day 9 after SCT, group 3-SCT control. All rats were investigated urodynamically on day 28 after transplantation. RESULTS BMSCs identified by BrdU immunohistochemistry survived in the injured spinal cord and lumbar level 3-4 (L(3-4)). Voiding pressure, episodes of non-voiding contractions and residual urine volumes were significantly decreased in BMSC rats, compared with the controls. Bladder capacity was similar in both groups. In four out of eight BMSC rats and one out of seven controls, the tonic and bursting external urethral sphincter electromyographic activity were detected during cystometry. Silent periods during bursting were shorter and activity periods were longer in BMSC rats compared with sham rats. CONCLUSION Intravenously transplanted BMSCs survived in the L(3-4) and had beneficial effects on the recovery of LUT function in the rats after SCT.
Collapse
|
47
|
The use of cellular magnetic resonance imaging to track the fate of iron-labeled multipotent stromal cells after direct transplantation in a mouse model of spinal cord injury. Mol Imaging Biol 2011; 13:702-11. [PMID: 20686855 DOI: 10.1007/s11307-010-0393-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE The objective of this study was to track the fate of iron-labeled, multipotent stromal cells (MSC) after their direct transplantation into mice with spinal cord injuries using magnetic resonance imaging (MRI). PROCEDURES Mice with spinal cord injuries received a direct transplant of (1) live MSC labeled with micron-sized iron oxide particles (MPIO); (2) dead, MPIO-labeled MSC; (3) unlabeled MSC; or (4) free MPIO and were imaged at 3 T for 6 weeks after transplantation. RESULTS Live, iron-labeled MSC appeared as a well-defined region of signal loss in the mouse spinal cord at the site of transplant. However, the MR appearance of dead, iron-labeled MSC and free iron particles was similar and persisted for the 6 weeks of the study. CONCLUSIONS Iron-labeled stem cells can be detected and monitored in vivo after direct transplantation into the injured spinal cord of mice. However, the fate of the iron label is not clear. Our investigation indicates that caution should be taken when interpreting MR images after direct transplantation of iron-labeled cells.
Collapse
|
48
|
Abstract
More than 1 million people in the United States live with a spinal cord injury (SCI). Despite medical advances, many patients with SCIs still experience substantial neurological disability, with loss of motor, sensory, and autonomic function. Cell therapy is ideally suited to address the multifactorial nature of the secondary events following SCI. Remarkable advances in our understanding of the pathophysiology of SCI, structural and functional magnetic resonance imaging, image-guided micro-neurosurgical techniques, and transplantable cell biology have enabled the use of cell-based regenerative techniques in the clinic. It is important to note that there are more than a dozen recently completed, ongoing, or recruiting cell therapy clinical trials for SCI that reflect the views of many key stakeholders. The field of regenerative neuroscience has reached a stage in which the clinical trials are scientifically and ethically justified. Although experimental models and analysis methods and techniques continue to evolve, no model will completely replicate the human condition. It is recognized that more work with cervical models of contusive/compressive SCI are required in parallel with clinical trials. It is also important that the clinical translation of advances made through well-established and validated experimental approaches in animal models move forward to meet the compelling needs of individuals with SCI and to advance the field of regenerative neuroscience. However, it is imperative that such efforts at translation be done in the most rigorous and informed fashion to determine safety and possible efficacy, and to provide key information to clinicians and basic scientists, which will allow improvements in regenerative techniques and the validation and refinement of existing preclinical animal models and research approaches. The field of regenerative neuroscience should not be stalled at the animal model stage, but instead the clinical trials need to be focused, safe, and ethical, backed up by a robust, translationally relevant preclinical research strategy.
Collapse
Affiliation(s)
- Michael G. Fehlings
- University Health Network, Toronto Western Hospital, Toronto, ON M5T 2S8 Canada
| | - Reaz Vawda
- University Health Network, Toronto Western Hospital, Toronto, ON M5T 2S8 Canada
| |
Collapse
|
49
|
Kikuchi K, Uchikado H, Miura N, Morimoto Y, Ito T, Tancharoen S, Miyata K, Sakamoto R, Kikuchi C, Iida N, Shiomi N, Kuramoto T, Miyagi N, Kawahara KI. HMGB1 as a therapeutic target in spinal cord injury: A hypothesis for novel therapy development. Exp Ther Med 2011; 2:767-770. [PMID: 22977572 DOI: 10.3892/etm.2011.310] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Accepted: 06/29/2011] [Indexed: 12/20/2022] Open
Abstract
Historically, clinical outcomes following spinal cord injury (SCI) have been dismal. Severe SCI leads to devastating neurological deficits, and there is no treatment available that restores the injury-induced loss of function to a degree that an independent life can be guaranteed. To address all the issues associated with SCI, a multidisciplinary approach is required, as it is unlikely that a single approach, such as surgical intervention, pharmacotherapy or cellular transplantation, will suffice. High mobility group box 1 (HMGB1) is an inflammatory cytokine. Various studies have shown that HMGB1 plays a critical role in SCI and that inhibition of HMGB1 release may be a novel therapeutic target for SCI and may support spinal cord repair. In addition, HMGB1 has been associated with graft rejection in the early phase. Therefore, HMGB1 may be a promising therapeutic target for SCI transplant patients. We hypothesize that inhibition of HMGB1 release rescues patients with SCI. Taken together, our findings suggest that anti-HMGB1 monoclonal antibodies or short hairpin RNA-mediated HMGB1 could be administered for spinal cord repair in SCI patients.
Collapse
Affiliation(s)
- Kiyoshi Kikuchi
- Department of Neurosurgery, Yame Public General Hospital, Yame 834-0034
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Motor-evoked potential confirmation of functional improvement by transplanted bone marrow mesenchymal stem cell in the ischemic rat brain. J Biomed Biotechnol 2011; 2011:238409. [PMID: 21772790 PMCID: PMC3134108 DOI: 10.1155/2011/238409] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2011] [Revised: 03/21/2011] [Accepted: 03/25/2011] [Indexed: 11/26/2022] Open
Abstract
This study investigated the effect of bone marrow mesenchymal stem cells (BMSCs) on the motor pathway in the transient ischemic rat brain that were transplanted through the carotid artery, measuring motor-evoked potential (MEP) in the four limbs muscle and the atlantooccipital membrane, which was elicited after monopolar and bipolar transcortical stimulation. After monopolar stimulation, the latency of MEP was significantly prolonged, and the amplitude was less reduced in the BMSC group in comparison with the control group (P < .05). MEPs induced by bipolar stimulation in the left forelimb could be measured in 40% of the BMSC group and the I wave that was not detected in the control group was also detected in 40% of the BMSC group. Our preliminary results imply that BMSCs transplanted to the ischemic rat brain mediate effects on the functional recovery of the cerebral motor cortex and the motor pathway.
Collapse
|