1
|
Zhang C, Morozova AY, Abakumov MA, Mel'nikov PA, Gabashvili AN, Chekhonin VP. Evaluation of the Optimal Number of Implanted Mesenchymal Stem Cells for the Treatment of Post-Traumatic Syrinx and Recovery of Motor Activity after Chronic Spinal Cord Injury. Bull Exp Biol Med 2023; 175:557-568. [PMID: 37773573 DOI: 10.1007/s10517-023-05904-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Indexed: 10/01/2023]
Abstract
The present work aims at determining the most effective dose (number) of mesenchymal stem cells (MSC) for its transplantation in order to treat chronic spinal cord injury (SCI) in mature Sprague-Dawley rats (n=24). MSC were obtained from bone marrow of 4-6-month-old Sprague-Dawley rats. Four weeks after SCI, MSC suspension (4 μl) was injected to experimental animals into the injured area in doses of 4×105, 8×105, or 106. Using MRI, diffusion tensor imaging (DTI), diffusion tensor tractography (DTT), immunohistochemistry, histological staining, and behavioral tests, we studied the effect of transplantation of MSC in different doses on the following parameters in rats with SCI: the size of lesion cavity and post-traumatic syrinx (PTS), glial scar formation, neuronal fibers remodeling, axonal regeneration and sprouting, vascularization, expression of neuronal factors, and motor functions. MSC administration improved motor function in rats after SCI due to stimulation of regeneration and sprouting of the axons, enhanced recovery of locomotor functions, reduction of PTS and the glial scar, and stimulation of vascularization and expression of the neurotrophic factors. The effects of MSC were dose-dependent; the most effective dose was 106 cells.
Collapse
Affiliation(s)
- C Zhang
- Department of Medicinal Nanobiotechnology, Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia.
- Department of Basic and Applied Neurobiology, V. P. Serbsky National Medical Research Center for Psychiatry and Narcology, Ministry of Health of the Russian Federation, Moscow, Russia.
| | - A Yu Morozova
- Department of Basic and Applied Neurobiology, V. P. Serbsky National Medical Research Center for Psychiatry and Narcology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - M A Abakumov
- Department of Medicinal Nanobiotechnology, Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia
| | - P A Mel'nikov
- Department of Medicinal Nanobiotechnology, Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia
| | - A N Gabashvili
- Department of Medicinal Nanobiotechnology, Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia
| | - V P Chekhonin
- Department of Medicinal Nanobiotechnology, Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia
- Department of Basic and Applied Neurobiology, V. P. Serbsky National Medical Research Center for Psychiatry and Narcology, Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
2
|
Zhang J, Steven ZD, Liao K. Therapeutic effect of umbilical cord blood cells on spinal cord injury. IBRAIN 2023; 9:195-204. [PMID: 37786552 PMCID: PMC10529013 DOI: 10.1002/ibra.12101] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 04/06/2023] [Accepted: 04/12/2023] [Indexed: 10/04/2023]
Abstract
Spinal cord injury (SCI) is a nervous system disease characterized by sensory and motor dysfunction, axonal apoptosis, decreased vascular density, and inflammation. At present, surgical treatment, drug treatment, and cell therapy can be used. Surgical treatment can improve motor and independent function scores, and drug treatment can promote the recovery of neurons in the spinal cord, but only improve symptoms. Complete recovery of SCI has not yet been achieved. However, the differentiation of stem cells brings hope for the treatment of SCI. Umbilical cord blood cells (UCBs) are ethically readily available and can repair neuronal damage. However, it is still unclear how they can improve symptoms and repair nerve severity. In this paper, the role of UCBs in the treatment of SCI is described in detail from different aspects such as behavior, morphology, and molecular expression changes, so as to provide new ideas and theoretical directions for future research.
Collapse
Affiliation(s)
- Jun‐Yan Zhang
- Department of AnesthesiologySouthwest Medical UniversityLuzhouChina
| | - Z. Du Steven
- Department of Integrative BiologyUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Ke‐Hua Liao
- The Sixth People's Hospital of ChengduChengduChina
| |
Collapse
|
3
|
Shi Y, Liu Y, Zhang B, Li X, Lin J, Yang C. Human Menstrual Blood-Derived Endometrial Stem Cells Promote Functional Recovery by Improving the Inflammatory Microenvironment in a Mouse Spinal Cord Injury Model. Cell Transplant 2023; 32:9636897231154579. [PMID: 36786359 PMCID: PMC9932767 DOI: 10.1177/09636897231154579] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023] Open
Abstract
Spinal cord injury (SCI) is a traumatic injury of the central nervous system. Because neurons are damaged and difficult to regenerate after SCI, its repair remains challenging. However, recent research on stem cell therapy have favored its use after SCI. In this study, based on the establishment of a mouse SCI model, human menstrual blood-derived endometrial stem cells (MenSCs) were intrathecally injected to explore the role and molecular mechanism of MenSCs in SCI. MenSCs were transplanted following SCI in the animal model, and behavioral evaluations showed that MenSC transplantation improved functional recovery. Therefore, samples were collected after 7 days, and transcriptome sequencing was performed. Gene Ontology (GO) enrichment analysis revealed that SCI is closely related to immune system processes. After transplantation of MenSCs, the immune response was significantly activated. In the Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, MenSC transplantation was found to be closely related to Th1, Th2, and Th17 cell differentiation pathways. Neuronal damage and glial cell proliferation and activation in the different groups were detected by fluorescence immunohistochemistry and Western blotting 7 days after SCI. Simultaneously, the activation of different types of microglia was detected and the expression of pro-inflammatory and anti-inflammatory factors was quantitatively analyzed. The results showed that MenSC transplantation and sonic hedgehog (Shh)-induced MenSCs accelerated neuronal recovery at the injured site, inhibited the formation of glial cells and microglial activation at the injured site, inhibited the expression of inflammatory factors, and improved the inflammatory microenvironment to achieve functional recovery of SCI. This study provides an experimental basis for the study of the role and molecular mechanism of MenSCs in SCI repair, and a reference for the role of Shh-induced MenSCs in SCI repair.
Collapse
Affiliation(s)
- Yaping Shi
- Stem Cells and Biotherapy Engineering
Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and
Biotherapy, School of Life Science and Technology, Xinxiang Medical University,
Xinxiang, China
| | - Yunfei Liu
- Stem Cells and Biotherapy Engineering
Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and
Biotherapy, School of Life Science and Technology, Xinxiang Medical University,
Xinxiang, China
| | - Bichao Zhang
- Stem Cells and Biotherapy Engineering
Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and
Biotherapy, School of Life Science and Technology, Xinxiang Medical University,
Xinxiang, China
| | - Xiaoying Li
- Stem Cells and Biotherapy Engineering
Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and
Biotherapy, School of Life Science and Technology, Xinxiang Medical University,
Xinxiang, China
| | - Juntang Lin
- Stem Cells and Biotherapy Engineering
Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and
Biotherapy, School of Life Science and Technology, Xinxiang Medical University,
Xinxiang, China,Henan Key Laboratory of Medical Tissue
Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Ciqing Yang
- Stem Cells and Biotherapy Engineering
Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and
Biotherapy, School of Life Science and Technology, Xinxiang Medical University,
Xinxiang, China,Henan Key Laboratory of Medical Tissue
Regeneration, Xinxiang Medical University, Xinxiang, China,Henan Key Laboratory of
Neurorestoratology, The First Affiliated Hospital of Xinxiang Medical University,
Xinxiang, China,Ciqing Yang, Stem Cells and Biotherapy
Engineering Research Center of Henan, National Joint Engineering Laboratory of
Stem Cells and Biotherapy, School of Life Science and Technology, Xinxiang
Medical University, Xinxiang 453003, China.
| |
Collapse
|
4
|
Roolfs L, Hubertus V, Spinnen J, Shopperly LK, Fehlings MG, Vajkoczy P. Therapeutic Approaches Targeting Vascular Repair After Experimental Spinal Cord Injury: A Systematic Review of the Literature. Neurospine 2022; 19:961-975. [PMID: 36597633 PMCID: PMC9816606 DOI: 10.14245/ns.2244624.312] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 09/16/2022] [Indexed: 12/27/2022] Open
Abstract
Traumatic spinal cord injury (SCI) disrupts the spinal cord vasculature resulting in ischemia, amplification of the secondary injury cascade and exacerbation of neural tissue loss. Restoring functional integrity of the microvasculature to prevent neural loss and to promote neural repair is an important challenge and opportunity in SCI research. Herein, we summarize the course of vascular injury and repair following SCI and give a comprehensive overview of current experimental therapeutic approaches targeting spinal cord microvasculature to diminish ischemia and thereby facilitate neural repair and regeneration. A systematic review of the published literature on therapeutic approaches to promote vascular repair after experimental SCI was performed using PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) standards. The MEDLINE databases PubMed, Embase, and OVID MEDLINE were searched using the keywords "spinal cord injury," "angiogenesis," "angiogenesis inducing agents," "tissue engineering," and "rodent subjects." A total of 111 studies were identified through the search. Five main therapeutic approaches to diminish hypoxia-ischemia and promote vascular repair were identified as (1) the application of angiogenic factors, (2) genetic engineering, (3) physical stimulation, (4) cell transplantation, and (5) biomaterials carrying various factor delivery. There are different therapeutic approaches with the potential to diminish hypoxia-ischemia and promote vascular repair after experimental SCI. Of note, combinatorial approaches using implanted biomaterials and angiogenic factor delivery appear promising for clinical translation.
Collapse
Affiliation(s)
- Laurens Roolfs
- Department of Neurosurgery, Charité – Universitätsmedizin Berlin and Berlin Institute of Health, Berlin, Germany
| | - Vanessa Hubertus
- Department of Neurosurgery, Charité – Universitätsmedizin Berlin and Berlin Institute of Health, Berlin, Germany
| | - Jacob Spinnen
- Tissue Engineering Laboratory, Charité – Universitätsmedizin Berlin and Berlin Institute of Health, Berlin, Germany
| | - Lennard K. Shopperly
- Tissue Engineering Laboratory, Charité – Universitätsmedizin Berlin and Berlin Institute of Health, Berlin, Germany
| | - Michael G. Fehlings
- Division of Neurosurgery and Krembil Neuroscience Centre, Toronto Western Hospital, University Health Network and University of Toronto, Toronto, Canada
| | - Peter Vajkoczy
- Department of Neurosurgery, Charité – Universitätsmedizin Berlin and Berlin Institute of Health, Berlin, Germany,Corresponding Author Peter Vajkoczy Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| |
Collapse
|
5
|
François I, Lepage OM, Carpenter E, Desjardins I, De Guio C, Benedetti ICC, Maddens S, Saulnier N, Grant BD. Mesenchymal stem cell transplantation into the spinal cord of healthy adult horses undergoing cervical ventral interbody fusion. Vet Surg 2021; 50:1107-1116. [PMID: 33709467 DOI: 10.1111/vsu.13611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 01/24/2021] [Accepted: 01/31/2021] [Indexed: 11/27/2022]
Abstract
OBJECTIVE To determine the feasibility of umbilical cord-derived mesenchymal stem cell (UC-MSC) transplantation into the cervical spinal cord of horses by using fluoroscopy with or without endoscopic guidance and to evaluate the neurological signs and tissue reaction after injection. STUDY DESIGN Experimental study. ANIMALS Eight healthy adult horses with no clinical signs of neurological disease. METHODS After cervical ventral interbody fusion (CVIF), ten million fluorescently labeled allogeneic UC-MSC were injected into the spinal cord under endoscopic and fluoroscopic guidance (n = 5) or fluoroscopic guidance only (n = 3). Postoperative neurological examinations were performed, and horses were humanely killed 48 hours (n = 4) or 14 days (n = 4) postoperatively. Spinal tissues were examined after gross dissection and with bright field and fluorescent microscopy. RESULTS Needle endoscopy of the cervical canal by ventral approach was associated with intraoperative spinal cord puncture (2/5) and postoperative ataxia (3/5). No intraoperative complications occurred, and one (1/3) horse developed ataxia with cell transplantation under fluoroscopy alone. Umbilical cord-derived MSC were associated with small vessels and detected up to 14 days in the spinal cord. Demyelination was observed in six of eight cases. CONCLUSION Fluoroscopically guided intramedullary UC-MSC transplantation during CVIF avoids spinal cord trauma and decreases risk of ataxia from endoscopy. Umbilical cord-derived MSC persist in the spinal cord for up to 14 days. Cell injection promotes angiogenesis and induces demyelination of the spinal tissue. CLINICAL SIGNIFICANCE Umbilical cord-derived MSC transplantation into the spinal cord during CVIF without endoscopy is recommended for future evaluation of cell therapy in horses affected by cervical vertebral compressive myelopathy.
Collapse
Affiliation(s)
- Isé François
- University of Lyon, VetAgro Sup, Veterinary Campus of Lyon, GREMERES-ICE Lyon Equine Research Center, France
| | - Olivier M Lepage
- University of Lyon, VetAgro Sup, Veterinary Campus of Lyon, GREMERES-ICE Lyon Equine Research Center, France
| | - Elaine Carpenter
- Cave Creek Equine Surgical and Diagnostic Imaging Center, Phoenix, Arizona, United States
| | - Isabelle Desjardins
- University of Lyon, VetAgro Sup, Veterinary Campus of Lyon, GREMERES-ICE Lyon Equine Research Center, France
| | - Cécile De Guio
- University of Lyon, VetAgro Sup, Veterinary Campus of Lyon, GREMERES-ICE Lyon Equine Research Center, France
| | | | | | | | | |
Collapse
|
6
|
Alishahi M, Anbiyaiee A, Farzaneh M, Khoshnam SE. Human Mesenchymal Stem Cells for Spinal Cord Injury. Curr Stem Cell Res Ther 2021; 15:340-348. [PMID: 32178619 DOI: 10.2174/1574888x15666200316164051] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 08/03/2019] [Accepted: 09/17/2019] [Indexed: 12/13/2022]
Abstract
Spinal Cord Injury (SCI), as a devastating and life-altering neurological disorder, is one of the most serious health issues. Currently, the management of acute SCI includes pharmacotherapy and surgical decompression. Both the approaches have been observed to have adverse physiological effects on SCI patients. Therefore, novel therapeutic targets for the management of SCI are urgently required for developing cell-based therapies. Multipotent stem cells, as a novel strategy for the treatment of tissue injury, may provide an effective therapeutic option against many neurological disorders. Mesenchymal stem cells (MSCs) or multipotent stromal cells can typically self-renew and generate various cell types. These cells are often isolated from bone marrow (BM-MSCs), adipose tissues (AD-MSCs), umbilical cord blood (UCB-MSCs), and placenta (PMSCs). MSCs have remarkable potential for the development of regenerative therapies in animal models and humans with SCI. Herein, we summarize the therapeutic potential of human MSCs in the treatment of SCI.
Collapse
Affiliation(s)
- Masoumeh Alishahi
- Department of Biology, Tehran North Branch, Islamic Azad University, Tehran, Iran
| | - Amir Anbiyaiee
- Department of Surgery, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz 61357-15794, Iran
| | - Maryam Farzaneh
- Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyed E Khoshnam
- Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
7
|
Moinuddin FM, Yolcu YU, Wahood W, Siddiqui AM, Chen BK, Alvi MA, Goyal A, Nesbitt JJ, Windebank AJ, Yeh JC, Petrucci K, Bydon M. Early and sustained improvements in motor function in rats after infusion of allogeneic umbilical cord-derived mesenchymal stem cells following spinal cord injury. Spinal Cord 2020; 59:319-327. [PMID: 33139846 DOI: 10.1038/s41393-020-00571-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 10/14/2020] [Accepted: 10/19/2020] [Indexed: 11/09/2022]
Abstract
STUDY DESIGN Animal study. OBJECTIVES Umbilical cord-derived mesenchymal stem cells (UC-MSCs) have recently been shown to hold great therapeutic potential for spinal cord injury (SCI). However, majority of the studies have been done using human cells transplanted into the rat with immunosuppression; this may not represent the outcomes that occur in humans. Herein, we present the therapeutic effect of using rat UC-MSCs (rUC-MSC) without immunosuppression in a rat model of SCI. SETTING Mayo Clinic, Rochester, MN, USA. METHODS Twelve female rats were randomly divided into two groups, control, and rUC-MSC group, and then subjected to a T9 moderate contusion SCI. Next, 2 × 106 rUC-MSCs or ringer-lactate solution were injected through the tail vein at 7 days post injury. Rats were assessed for 14 weeks by an open-field Basso, Beattie, and Bresnahan (BBB) motor score as well as postmortem quantification of axonal sparing/regeneration, cavity volume, and glial scar. RESULTS Animals treated with rUC-MSCs were found to have early and sustained motor improvement (BBB score of 14.6 ± 1.9 compared to 10.1 ± 1.7 in the control group) at 14 weeks post injury (mean difference: 4.55, 95% CI: 2.04 to 7.06; p value < 0.001). Total cavity volume in the injury epicenter was significantly reduced in the rUC-MSC group; control: 33.0% ± 2.1, rUC-MSC: 25.3% ± 3.8 (mean difference: -7.7% (95% CI: -12.3 to -2.98); p value < 0.05). In addition, spinal cords from rats treated with rUC-MSCs were found to have a significantly greater number of myelinated axons, decreased astrogliosis, and reduced glial scar formation compared to control rats. CONCLUSIONS Our study indicates that intravenous injection of allogenic UC-MSCs without immunosuppression exert beneficial effects in subacute SCI and thus could be a useful therapy to improve the functional capacity among patients with SCI.
Collapse
Affiliation(s)
- F M Moinuddin
- Mayo Clinic Neuro-Informatics Laboratory, Mayo Clinic, Rochester, MN, USA.,Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Yagiz U Yolcu
- Mayo Clinic Neuro-Informatics Laboratory, Mayo Clinic, Rochester, MN, USA.,Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Waseem Wahood
- Mayo Clinic Neuro-Informatics Laboratory, Mayo Clinic, Rochester, MN, USA.,Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
| | | | - Bingkun K Chen
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Mohammed Ali Alvi
- Mayo Clinic Neuro-Informatics Laboratory, Mayo Clinic, Rochester, MN, USA.,Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Anshit Goyal
- Mayo Clinic Neuro-Informatics Laboratory, Mayo Clinic, Rochester, MN, USA.,Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | | - Mohamad Bydon
- Mayo Clinic Neuro-Informatics Laboratory, Mayo Clinic, Rochester, MN, USA. .,Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
8
|
Jin MC, Medress ZA, Azad TD, Doulames VM, Veeravagu A. Stem cell therapies for acute spinal cord injury in humans: a review. Neurosurg Focus 2020; 46:E10. [PMID: 30835679 DOI: 10.3171/2018.12.focus18602] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 12/11/2018] [Indexed: 12/21/2022]
Abstract
Recent advances in stem cell biology present significant opportunities to advance clinical applications of stem cell-based therapies for spinal cord injury (SCI). In this review, the authors critically analyze the basic science and translational evidence that supports the use of various stem cell sources, including induced pluripotent stem cells, oligodendrocyte precursor cells, and mesenchymal stem cells. They subsequently explore recent advances in stem cell biology and discuss ongoing clinical translation efforts, including combinatorial strategies utilizing scaffolds, biogels, and growth factors to augment stem cell survival, function, and engraftment. Finally, the authors discuss the evolution of stem cell therapies for SCI by providing an overview of completed (n = 18) and ongoing (n = 9) clinical trials.
Collapse
|
9
|
Lee S, Prisby RD. Short-term intermittent parathyroid hormone (1-34) administration increased angiogenesis and matrix metalloproteinase 9 in femora of mature and middle-aged C57BL/6 mice. Exp Physiol 2020; 105:1159-1171. [PMID: 32306445 DOI: 10.1113/ep087869] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 04/15/2020] [Indexed: 01/09/2023]
Abstract
NEW FINDINGS What is the central question of this study? We sought to assess the effects of intermittent parathyroid hormone (1-34) administration on bone angiogenesis, the redistribution of bone marrow blood vessels, and matrix metalloproteinase 9 as a function of advancing age in mice. What is the main finding and its importance? Short-term (i.e. 10 days) intermittent parathyroid hormone (1-34) administration increased the number of small (≤29-µm-diameter) bone marrow blood vessels and augmented matrix metalloproteinase 9. These changes occurred before alterations in trabecular bone. Given the rapid response in bone angiogenesis, this investigation highlights the impact of intermittent parathyroid hormone (1-34) administration on the bone vascular network. ABSTRACT Intermittent parathyroid hormone (PTH) administration augments bone, stimulates the production of matrix metalloproteinase 9 (Mmp9) and relocates bone marrow blood vessels closer to osteoid seams. Discrepancies exist, however, regarding bone angiogenesis. Given that Mmp9 participates in cellular homing and migration, it might aid in blood vessel relocation. We examined the influence of short-term intermittent PTH administration on angiogenesis, Mmp9 secretion and the distance between blood vessels and bone. Mature (6- to 8-month-old) and middle-aged (10- to 12-month-old) male and female C57BL/6 mice were divided into three groups: control (CON), and 5 (5dPTH) and 10 days (10dPTH) of intermittent PTH administration. Mice were given PBS (50 µl day-1 ) or PTH(1-34) (43 µg kg-1 day-1 ). Frontal sections (5 µm thick) of the right distal femoral metaphysis were triple-immunolabelled to identify endothelial cells (anti-CD31), vascular smooth muscle cells (anti-αSMA) and Mmp9 (anti-Mmp9). Vascular density, Mmp9 density, area and localization, and blood vessel distance from bone were analysed. Blood vessels were analysed according to diameter: 1-29, 30-100 and 101-200 µm. Trabecular bone microarchitecture and bone static and dynamic properties were assessed. No main effects of age were observed for any variable. The density of CD31-labelled blood vessels 1-29 and 30-100 µm in diameter was higher (P < 0.05) and tended (P = 0.055) to be higher, respectively, in 10dPTH versus 5dPTH and CON. Mmp9 was augmented (P < 0.05) in 10dPTH versus the other groups. Mmp9 was closer (P < 0.05) to blood vessels 1-29 µm in diameter and furthest (P < 0.05) from bone. In conclusion, bone angiogenesis occurred by day 10 of intermittent PTH administration, coinciding with augmented Mmp9 secretion near the smallest blood vessels (1-29 µm in diameter).
Collapse
Affiliation(s)
- Seungyong Lee
- Department of Kinesiology, University of Texas at Arlington, Arlington, TX, USA
| | - Rhonda D Prisby
- Department of Kinesiology, University of Texas at Arlington, Arlington, TX, USA
| |
Collapse
|
10
|
Dalamagkas K, Tsintou M, Seifalian A, Seifalian AM. Translational Regenerative Therapies for Chronic Spinal Cord Injury. Int J Mol Sci 2018; 19:E1776. [PMID: 29914060 PMCID: PMC6032191 DOI: 10.3390/ijms19061776] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 06/05/2018] [Accepted: 06/06/2018] [Indexed: 12/22/2022] Open
Abstract
Spinal cord injury is a chronic and debilitating neurological condition that is currently being managed symptomatically with no real therapeutic strategies available. Even though there is no consensus on the best time to start interventions, the chronic phase is definitely the most stable target in order to determine whether a therapy can effectively restore neurological function. The advancements of nanoscience and stem cell technology, combined with the powerful, novel neuroimaging modalities that have arisen can now accelerate the path of promising novel therapeutic strategies from bench to bedside. Several types of stem cells have reached up to clinical trials phase II, including adult neural stem cells, human spinal cord stem cells, olfactory ensheathing cells, autologous Schwann cells, umbilical cord blood-derived mononuclear cells, adult mesenchymal cells, and autologous bone-marrow-derived stem cells. There also have been combinations of different molecular therapies; these have been either alone or combined with supportive scaffolds with nanostructures to facilitate favorable cell⁻material interactions. The results already show promise but it will take some coordinated actions in order to develop a proper step-by-step approach to solve impactful problems with neural repair.
Collapse
Affiliation(s)
- Kyriakos Dalamagkas
- The Institute for Rehabilitation and Research, Memorial Hermann Texas Medical Centre, Houston, TX 77030, USA.
- Centre for Nanotechnology & Regenerative Medicine, Division of Surgery and Interventional Science, University College of London (UCL), London NW3 2QG, UK.
| | - Magdalini Tsintou
- Centre for Nanotechnology & Regenerative Medicine, Division of Surgery and Interventional Science, University College of London (UCL), London NW3 2QG, UK.
- Center for Neural Systems Investigations, Massachusetts General Hospital/HST Athinoula A., Martinos Centre for Biomedical Imaging, Harvard Medical School, Boston, MA 02129, USA.
| | - Amelia Seifalian
- Faculty of Medical Sciences, UCL Medical School, London WC1E 6BT, UK.
| | - Alexander M Seifalian
- NanoRegMed Ltd. (Nanotechnology & Regenerative Medicine Commercialization Centre), The London BioScience Innovation Centre, London NW1 0NH, UK.
| |
Collapse
|
11
|
Zhu H, Poon W, Liu Y, Leung GKK, Wong Y, Feng Y, Ng SCP, Tsang KS, Sun DTF, Yeung DK, Shen C, Niu F, Xu Z, Tan P, Tang S, Gao H, Cha Y, So KF, Fleischaker R, Sun D, Chen J, Lai J, Cheng W, Young W. Phase I-II Clinical Trial Assessing Safety and Efficacy of Umbilical Cord Blood Mononuclear Cell Transplant Therapy of Chronic Complete Spinal Cord Injury. Cell Transplant 2018; 25:1925-1943. [PMID: 27075659 DOI: 10.3727/096368916x691411] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Umbilical cord blood-derived mononuclear cell (UCB-MNC) transplants improve recovery in animal spinal cord injury (SCI) models. We transplanted UCB-MNCs into 28 patients with chronic complete SCI in Hong Kong (HK) and Kunming (KM). Stemcyte Inc. donated UCB-MNCs isolated from human leukocyte antigen (HLA ≥4:6)-matched UCB units. In HK, four patients received four 4-μl injections (1.6 million cells) into dorsal entry zones above and below the injury site, and another four received 8-μl injections (3.2 million cells). The eight patients were an average of 13 years after C5-T10 SCI. Magnetic resonance diffusion tensor imaging of five patients showed white matter gaps at the injury site before treatment. Two patients had fiber bundles growing across the injury site by 12 months, and the rest had narrower white matter gaps. Motor, walking index of SCI (WISCI), and spinal cord independence measure (SCIM) scores did not change. In KM, five groups of four patients received four 4-μl (1.6 million cells), 8-μl (3.2 million cells), 16-μl injections (6.4 million cells), 6.4 million cells plus 30 mg/kg methylprednisolone (MP), or 6.4 million cells plus MP and a 6-week course of oral lithium carbonate (750 mg/day). KM patients averaged 7 years after C3-T11 SCI and received 3-6 months of intensive locomotor training. Before surgery, only two patients walked 10 m with assistance and did not need assistance for bladder or bowel management before surgery. The rest could not walk or do their bladder and bowel management without assistance. At about a year (41-87 weeks), WISCI and SCIM scores improved: 15/20 patients walked 10 m ( p = 0.001) and 12/20 did not need assistance for bladder management ( p = 0.001) or bowel management ( p = 0.002). Five patients converted from complete to incomplete (two sensory, three motor; p = 0.038) SCI. We conclude that UCB-MNC transplants and locomotor training improved WISCI and SCIM scores. We propose further clinical trials.
Collapse
Affiliation(s)
- Hui Zhu
- Kunming General Hospital of Chengdu Military Command, Yunnan, P.R. China.,Kunming Tongren Hospital, Yunnan, P.R. China
| | - Waisang Poon
- Prince of Wales Hospital, Division of Neurosurgery, Department of Surgery, Chinese University of Hong Kong, Shatin, Hong Kong, SAR, P.R. China
| | - Yansheng Liu
- Kunming General Hospital of Chengdu Military Command, Yunnan, P.R. China.,Kunming Tongren Hospital, Yunnan, P.R. China
| | | | - Yatwa Wong
- Queen Mary Hospital, University of Hong Kong, Hong Kong, SAR, P.R. China
| | - Yaping Feng
- Kunming General Hospital of Chengdu Military Command, Yunnan, P.R. China
| | - Stephanie C P Ng
- Prince of Wales Hospital, Division of Neurosurgery, Department of Surgery, Chinese University of Hong Kong, Shatin, Hong Kong, SAR, P.R. China
| | - Kam Sze Tsang
- Prince of Wales Hospital, Division of Neurosurgery, Department of Surgery, Chinese University of Hong Kong, Shatin, Hong Kong, SAR, P.R. China
| | - David T F Sun
- Prince of Wales Hospital, Division of Neurosurgery, Department of Surgery, Chinese University of Hong Kong, Shatin, Hong Kong, SAR, P.R. China
| | - David K Yeung
- Prince of Wales Hospital, Division of Neurosurgery, Department of Surgery, Chinese University of Hong Kong, Shatin, Hong Kong, SAR, P.R. China
| | - Caihong Shen
- Kunming General Hospital of Chengdu Military Command, Yunnan, P.R. China.,Kunming Tongren Hospital, Yunnan, P.R. China
| | - Fang Niu
- Kunming General Hospital of Chengdu Military Command, Yunnan, P.R. China.,Kunming Tongren Hospital, Yunnan, P.R. China
| | - Zhexi Xu
- Kunming General Hospital of Chengdu Military Command, Yunnan, P.R. China.,Kunming Tongren Hospital, Yunnan, P.R. China
| | - Pengju Tan
- Kunming General Hospital of Chengdu Military Command, Yunnan, P.R. China.,Kunming Tongren Hospital, Yunnan, P.R. China
| | - Shaofeng Tang
- Kunming General Hospital of Chengdu Military Command, Yunnan, P.R. China
| | - Hongkun Gao
- Kunming General Hospital of Chengdu Military Command, Yunnan, P.R. China.,Kunming Tongren Hospital, Yunnan, P.R. China
| | - Yun Cha
- Kunming General Hospital of Chengdu Military Command, Yunnan, P.R. China
| | - Kwok-Fai So
- Department of Ophthalmology and State Key Laboratory of Brain and Cognitive Science, The University of Hong Kong, SAR, P.R. China.,GHM Institute of CNS Regeneration, and Medical Key Laboratory of Brain Function and Diseases, Jinan University, Guangzhou, P.R. China.,China Spinal Cord Injury Network, Hong Kong Science Technology Park, Hong Kong, SAR, P.R. China
| | | | - Dongming Sun
- W. M. Keck Center for Collaborative Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - John Chen
- China Spinal Cord Injury Network, Hong Kong Science Technology Park, Hong Kong, SAR, P.R. China
| | - Jan Lai
- China Spinal Cord Injury Network, Hong Kong Science Technology Park, Hong Kong, SAR, P.R. China
| | - Wendy Cheng
- China Spinal Cord Injury Network, Hong Kong Science Technology Park, Hong Kong, SAR, P.R. China
| | - Wise Young
- China Spinal Cord Injury Network, Hong Kong Science Technology Park, Hong Kong, SAR, P.R. China.,W. M. Keck Center for Collaborative Neuroscience, Rutgers University, Piscataway, NJ, USA
| |
Collapse
|
12
|
Chang HC. The role of policies and networks in development of cord blood usage in China. Regen Med 2017; 12:637-645. [DOI: 10.2217/rme-2017-0050] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Research regarding the use of cord blood (CB) has focused on antigen match and the number of stem cells, with policies and networks related to its use being under researched. This article is based on fieldwork in China from 2013 to 2015 and examines ways that the studied CB bank enhances CB usage in China. This article identifies that in addition to finding a match, CB use is linked to the policies and networks, release fee and public awareness that enable CB usage development.
Collapse
Affiliation(s)
- Hung-Chieh Chang
- Institute of Health Policy & Management, National Taiwan University No. 17, Xu-Zhou Road, Taipei 100, Taiwan
| |
Collapse
|
13
|
|
14
|
Zhang C, Feng S, Hu N, Douglas P, Chekhonin VP. Letter to the editor regarding "Local versus distal transplantation of human neural stem cells following chronic spinal cord injury" by Cheng et al. Spine J 2016; 16:792-3. [PMID: 27342709 DOI: 10.1016/j.spinee.2016.02.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 02/08/2016] [Indexed: 02/03/2023]
Affiliation(s)
- Chao Zhang
- Department of Orthopedics, Tianjin Medical University General Hospital, Anshan Rd 154th Heping Area, Tianjin 300070, China; Department of Medicinal Nanobiotechnology, Pirogov Russian National Research Medical University, Ostrovitianov str. 1, Moscow 117997, Russia; Department of Basic and Applied Neurobiology, Federal Medical Research Center for Psychiatry and Narcology, Kropotkinskiy Lane 23th, Moscow 119119, Russia
| | - Shiqing Feng
- Department of Orthopedics, Tianjin Medical University General Hospital, Anshan Rd 154th Heping Area, Tianjin 300070, China
| | - Nan Hu
- Department of Anesthesiology, Tianjin Medical University General Hospital, Anshan Rd 154th Heping Area, Tianjin 300070, China
| | - Patricia Douglas
- Department of Orthopedics, Tianjin Medical University General Hospital, Anshan Rd 154th Heping Area, Tianjin 300070, China
| | - Vladimir P Chekhonin
- Department of Medicinal Nanobiotechnology, Pirogov Russian National Research Medical University, Ostrovitianov str. 1, Moscow 117997, Russia; Department of Basic and Applied Neurobiology, Federal Medical Research Center for Psychiatry and Narcology, Kropotkinskiy Lane 23th, Moscow 119119, Russia
| |
Collapse
|
15
|
Gwak SJ, Yun Y, Yoon DH, Kim KN, Ha Y. Therapeutic Use of 3β-[N-(N',N'-Dimethylaminoethane) Carbamoyl] Cholesterol-Modified PLGA Nanospheres as Gene Delivery Vehicles for Spinal Cord Injury. PLoS One 2016; 11:e0147389. [PMID: 26824765 PMCID: PMC4732605 DOI: 10.1371/journal.pone.0147389] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 01/04/2016] [Indexed: 01/19/2023] Open
Abstract
Gene delivery holds therapeutic promise for the treatment of neurological diseases and spinal cord injury. Although several studies have investigated the use of non-viral vectors, such as polyethylenimine (PEI), their clinical value is limited by their cytotoxicity. Recently, biodegradable poly (lactide-co-glycolide) (PLGA) nanospheres have been explored as non-viral vectors. Here, we show that modification of PLGA nanospheres with 3β-[N-(N′,N′-dimethylaminoethane) carbamoyl] cholesterol (DC-Chol) enhances gene transfection efficiency. PLGA/DC-Chol nanospheres encapsulating DNA were prepared using a double emulsion-solvent evaporation method. PLGA/DC-Chol nanospheres were less cytotoxic than PEI both in vitro and in vivo. DC-Chol modification improved the uptake of nanospheres, thereby increasing their transfection efficiency in mouse neural stem cells in vitro and rat spinal cord in vivo. Also, transgene expression induced by PLGA nanospheres was higher and longer-lasting than that induced by PEI. In a rat model of spinal cord injury, PLGA/DC-Chol nanospheres loaded with vascular endothelial growth factor gene increased angiogenesis at the injury site, improved tissue regeneration, and resulted in better recovery of locomotor function. These results suggest that DC-Chol-modified PLGA nanospheres could serve as therapeutic gene delivery vehicles for spinal cord injury.
Collapse
Affiliation(s)
- So-Jung Gwak
- Spine & Spinal Cord Institute, Department of Neurosurgery, Yonsei University College of Medicine, Seoul, Korea
- Department of Bioengineering, Clemson University, Clemson, South Carolina, United States of America
| | - Yeomin Yun
- Spine & Spinal Cord Institute, Department of Neurosurgery, Yonsei University College of Medicine, Seoul, Korea
| | - Do Heum Yoon
- Spine & Spinal Cord Institute, Department of Neurosurgery, Yonsei University College of Medicine, Seoul, Korea
| | - Keung Nyun Kim
- Spine & Spinal Cord Institute, Department of Neurosurgery, Yonsei University College of Medicine, Seoul, Korea
| | - Yoon Ha
- Spine & Spinal Cord Institute, Department of Neurosurgery, Yonsei University College of Medicine, Seoul, Korea
- * E-mail:
| |
Collapse
|
16
|
Spinal cord injury repair by implantation of structured hyaluronic acid scaffold with PLGA microspheres in the rat. Cell Tissue Res 2015; 364:17-28. [DOI: 10.1007/s00441-015-2298-1] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 09/25/2015] [Indexed: 11/26/2022]
|
17
|
McMahill BG, Spriet M, Sisó S, Manzer MD, Mitchell G, McGee J, Garcia TC, Borjesson DL, Sieber-Blum M, Nolta JA, Sturges BK. Feasibility Study of Canine Epidermal Neural Crest Stem Cell Transplantation in the Spinal Cords of Dogs. Stem Cells Transl Med 2015; 4:1173-86. [PMID: 26273065 DOI: 10.5966/sctm.2015-0018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 06/17/2015] [Indexed: 01/12/2023] Open
Abstract
UNLABELLED This pilot feasibility study aimed to determine the outcome of canine epidermal neural crest stem cell (cEPI-NCSC) grafts in the normal spinal cords of healthy bred-for-research dogs. This included developing novel protocols for (a) the ex vivo expansion of cEPI-NCSCs, (b) the delivery of cEPI-NCSCs into the spinal cord, and (c) the labeling of the cells and subsequent tracing of the graft in the live animal by magnetic resonance imaging. A total of four million cEPI-NCSCs were injected into the spinal cord divided in two locations. Differences in locomotion at baseline and post-treatment were evaluated by gait analysis and compared with neurological outcome and behavioral exams. Histopathological analyses of the spinal cords and cEPI-NCSC grafts were performed at 3 weeks post-transplantation. Neurological and gait parameters were minimally affected by the stem cell injection. cEPI-NCSCs survived in the canine spinal cord for the entire period of investigation and did not migrate or proliferate. Subsets of cEPI-NCSCs expressed the neural crest stem cell marker Sox10. There was no detectable expression of markers for glial cells or neurons. The tissue reaction to the cell graft was predominantly vascular in addition to a degree of reactive astrogliosis and microglial activation. In the present study, we demonstrated that cEPI-NCSC grafts survive in the spinal cords of healthy dogs without major adverse effects. They persist locally in the normal spinal cord, may promote angiogenesis and tissue remodeling, and elicit a tissue response that may be beneficial in patients with spinal cord injury. SIGNIFICANCE It has been established that mouse and human epidermal neural crest stem cells are somatic multipotent stem cells with proved innovative potential in a mouse model of spinal cord injury (SCI) offering promise of a valid treatment for SCI. Traumatic SCI is a common neurological problem in dogs with marked similarities, clinically and pathologically, to the syndrome in people. For this reason, dogs provide a readily accessible, clinically realistic, spontaneous model for evaluation of epidermal neural crest stem cells therapeutic intervention. The results of this study are expected to give the baseline data for a future clinical trial in dogs with traumatic SCI.
Collapse
Affiliation(s)
- Barbara G McMahill
- Stem Cell Program, Institute for Regenerative Cures, University of California Davis Medical Center, Sacramento, California, USA; Department of Surgical and Radiological Sciences, Department of Pathology, Microbiology and Immunology, and J.D. Wheat Veterinary Orthopedic Research Laboratory, School of Veterinary Medicine, University of California, Davis, California, USA; Institute of Genetic Medicine, Centre for Life, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Mathieu Spriet
- Stem Cell Program, Institute for Regenerative Cures, University of California Davis Medical Center, Sacramento, California, USA; Department of Surgical and Radiological Sciences, Department of Pathology, Microbiology and Immunology, and J.D. Wheat Veterinary Orthopedic Research Laboratory, School of Veterinary Medicine, University of California, Davis, California, USA; Institute of Genetic Medicine, Centre for Life, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Sílvia Sisó
- Stem Cell Program, Institute for Regenerative Cures, University of California Davis Medical Center, Sacramento, California, USA; Department of Surgical and Radiological Sciences, Department of Pathology, Microbiology and Immunology, and J.D. Wheat Veterinary Orthopedic Research Laboratory, School of Veterinary Medicine, University of California, Davis, California, USA; Institute of Genetic Medicine, Centre for Life, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Michael D Manzer
- Stem Cell Program, Institute for Regenerative Cures, University of California Davis Medical Center, Sacramento, California, USA; Department of Surgical and Radiological Sciences, Department of Pathology, Microbiology and Immunology, and J.D. Wheat Veterinary Orthopedic Research Laboratory, School of Veterinary Medicine, University of California, Davis, California, USA; Institute of Genetic Medicine, Centre for Life, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Gaela Mitchell
- Stem Cell Program, Institute for Regenerative Cures, University of California Davis Medical Center, Sacramento, California, USA; Department of Surgical and Radiological Sciences, Department of Pathology, Microbiology and Immunology, and J.D. Wheat Veterinary Orthopedic Research Laboratory, School of Veterinary Medicine, University of California, Davis, California, USA; Institute of Genetic Medicine, Centre for Life, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Jeannine McGee
- Stem Cell Program, Institute for Regenerative Cures, University of California Davis Medical Center, Sacramento, California, USA; Department of Surgical and Radiological Sciences, Department of Pathology, Microbiology and Immunology, and J.D. Wheat Veterinary Orthopedic Research Laboratory, School of Veterinary Medicine, University of California, Davis, California, USA; Institute of Genetic Medicine, Centre for Life, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Tanya C Garcia
- Stem Cell Program, Institute for Regenerative Cures, University of California Davis Medical Center, Sacramento, California, USA; Department of Surgical and Radiological Sciences, Department of Pathology, Microbiology and Immunology, and J.D. Wheat Veterinary Orthopedic Research Laboratory, School of Veterinary Medicine, University of California, Davis, California, USA; Institute of Genetic Medicine, Centre for Life, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Dori L Borjesson
- Stem Cell Program, Institute for Regenerative Cures, University of California Davis Medical Center, Sacramento, California, USA; Department of Surgical and Radiological Sciences, Department of Pathology, Microbiology and Immunology, and J.D. Wheat Veterinary Orthopedic Research Laboratory, School of Veterinary Medicine, University of California, Davis, California, USA; Institute of Genetic Medicine, Centre for Life, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Maya Sieber-Blum
- Stem Cell Program, Institute for Regenerative Cures, University of California Davis Medical Center, Sacramento, California, USA; Department of Surgical and Radiological Sciences, Department of Pathology, Microbiology and Immunology, and J.D. Wheat Veterinary Orthopedic Research Laboratory, School of Veterinary Medicine, University of California, Davis, California, USA; Institute of Genetic Medicine, Centre for Life, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Jan A Nolta
- Stem Cell Program, Institute for Regenerative Cures, University of California Davis Medical Center, Sacramento, California, USA; Department of Surgical and Radiological Sciences, Department of Pathology, Microbiology and Immunology, and J.D. Wheat Veterinary Orthopedic Research Laboratory, School of Veterinary Medicine, University of California, Davis, California, USA; Institute of Genetic Medicine, Centre for Life, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Beverly K Sturges
- Stem Cell Program, Institute for Regenerative Cures, University of California Davis Medical Center, Sacramento, California, USA; Department of Surgical and Radiological Sciences, Department of Pathology, Microbiology and Immunology, and J.D. Wheat Veterinary Orthopedic Research Laboratory, School of Veterinary Medicine, University of California, Davis, California, USA; Institute of Genetic Medicine, Centre for Life, Newcastle University, Newcastle Upon Tyne, United Kingdom
| |
Collapse
|
18
|
Hu JZ, Long H, Wu TD, Zhou Y, Lu HB. The effect of estrogen-related receptor α on the regulation of angiogenesis after spinal cord injury. Neuroscience 2015; 290:570-80. [PMID: 25665753 DOI: 10.1016/j.neuroscience.2015.01.067] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 01/29/2015] [Accepted: 01/29/2015] [Indexed: 01/08/2023]
Abstract
Estrogen receptor-related receptor-α (ERRα) is an orphan member of the nuclear receptor superfamily that interacts with peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α) to stimulate vascular endothelial growth factor (VEGF) expression and angiogenesis in a hypoxia-inducible factor-1α-independent pathway. Although it is not regulated by any natural ligand, the action of ERRα can be blocked by the synthetic molecule XCT790. In the present study, Sprague-Dawley rats were randomly allocated to a sham group, injury-saline group or injury-XCT90 group. A modified Allen's weight-drop method was applied to induce the acute traumatic spinal cord injury (SCI) model in these rats, and an injection of XCT790 was administered every 24h, starting half an hour after the SCI contusion. Histological analyses revealed that XCT790 significantly aggravated tissue damage and decreased the number of ERRα-positive cells at 1, 3 and 7 days after SCI. Western blot and quantitative real-time polymerase chain reaction (qRT-PCR) analyses also indicated that XCT790 dramatically repressed the expression of ERRα, thus reducing the expression of VEGF and angiopoietin-2 (Ang-2) throughout the duration of the experiment, but the expression of PGC-1α was not affected. Immunofluorescence analyses indicated that vascular density and endothelial cell proliferation were decreased in the injury-XCT90 group compared with the injury-saline group. These results suggest that ERRα is involved in mediating angiogenesis after SCI in the rat traumatic SCI model.
Collapse
Affiliation(s)
- J Z Hu
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha 410008, PR China
| | - H Long
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha 410008, PR China
| | - T-D Wu
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha 410008, PR China
| | - Y Zhou
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha 410008, PR China
| | - H-B Lu
- Department of Sports Medicine, Research Center of Sports Medicine, Xiangya Hospital, Central South University, Changsha 410008, PR China.
| |
Collapse
|
19
|
Zhou H, Li X, Wu Q, Li F, Fu Z, Liu C, Liang Z, Chu T, Wang T, Lu L, Ning G, Kong X, Feng S. shRNA against PTEN promotes neurite outgrowth of cortical neurons and functional recovery in spinal cord contusion rats. Regen Med 2014; 10:411-29. [PMID: 25495396 DOI: 10.2217/rme.14.88] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
AIM To explore neurite growth/regeneration and spinal cord injury repair after PTEN silencing via lentivirus-mediated RNAi. MATERIALS & METHODS Cortical neurons were seeded on or adjacent to chondroitin sulfate proteoglycans. The length, number and crossing behavior of neurites were calculated. Lentivirus was locally injected into spinal cord contusion rats. The functional recovery and immunohistochemical staining were analyzed. RESULTS Neurites with PTEN silencing exhibited significant enhancements in elongation, initiation and crossing ability when they encountered chondroitin sulfate proteoglycans in vitro. In vivo PTEN silencing improved functional recovery significantly, and promoted axon and synapse formation, but not scar formation. CONCLUSIONS PTEN silencing may be promising for spinal cord injury repair.
Collapse
Affiliation(s)
- Hengxing Zhou
- 1Department of Orthopaedics, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin 300052, PR China
| | | | - Qiang Wu
- 3Department of Orthopaedics, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No. 314 Anshanxi Road, Nankai District, Tianjin 300193, PR China
| | - Fuyuan Li
- 1Department of Orthopaedics, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin 300052, PR China
| | | | - Chang Liu
- 4School of Medicine, Nankai University, No. 94 Weijin Road, Nankai District, Tianjin 300071, PR China
| | - Zhipin Liang
- 4School of Medicine, Nankai University, No. 94 Weijin Road, Nankai District, Tianjin 300071, PR China
| | - Tianci Chu
- 1Department of Orthopaedics, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin 300052, PR China
| | - Tianyi Wang
- 1Department of Orthopaedics, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin 300052, PR China
| | - Lu Lu
- 1Department of Orthopaedics, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin 300052, PR China
| | - Guangzhi Ning
- 1Department of Orthopaedics, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin 300052, PR China
| | - Xiaohong Kong
- 4School of Medicine, Nankai University, No. 94 Weijin Road, Nankai District, Tianjin 300071, PR China
| | - Shiqing Feng
- 1Department of Orthopaedics, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin 300052, PR China
| |
Collapse
|
20
|
Dasari VR, Veeravalli KK, Dinh DH. Mesenchymal stem cells in the treatment of spinal cord injuries: A review. World J Stem Cells 2014; 6:120-133. [PMID: 24772239 PMCID: PMC3999770 DOI: 10.4252/wjsc.v6.i2.120] [Citation(s) in RCA: 147] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 02/19/2014] [Accepted: 03/12/2014] [Indexed: 02/06/2023] Open
Abstract
With technological advances in basic research, the intricate mechanism of secondary delayed spinal cord injury (SCI) continues to unravel at a rapid pace. However, despite our deeper understanding of the molecular changes occurring after initial insult to the spinal cord, the cure for paralysis remains elusive. Current treatment of SCI is limited to early administration of high dose steroids to mitigate the harmful effect of cord edema that occurs after SCI and to reduce the cascade of secondary delayed SCI. Recent evident-based clinical studies have cast doubt on the clinical benefit of steroids in SCI and intense focus on stem cell-based therapy has yielded some encouraging results. An array of mesenchymal stem cells (MSCs) from various sources with novel and promising strategies are being developed to improve function after SCI. In this review, we briefly discuss the pathophysiology of spinal cord injuries and characteristics and the potential sources of MSCs that can be used in the treatment of SCI. We will discuss the progress of MSCs application in research, focusing on the neuroprotective properties of MSCs. Finally, we will discuss the results from preclinical and clinical trials involving stem cell-based therapy in SCI.
Collapse
|
21
|
Dedeepiya VD, William JB, Parthiban JKBC, Chidambaram R, Balamurugan M, Kuroda S, Iwasaki M, Preethy S, Abraham SJK. The known-unknowns in spinal cord injury, with emphasis on cell-based therapies - a review with suggestive arenas for research. Expert Opin Biol Ther 2014; 14:617-34. [PMID: 24660978 DOI: 10.1517/14712598.2014.889676] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION In spite of extensive research, the progress toward a cure in spinal cord injury (SCI) is still elusive, which holds good for the cell- and stem cell-based therapies. We have critically analyzed seven known gray areas in SCI, indicating the specific arenas for research to improvise the outcome of cell-based therapies in SCI. AREAS COVERED The seven, specific known gray areas in SCI analyzed are: i) the gap between animal models and human victims; ii) uncertainty about the time, route and dosage of cells applied; iii) source of the most efficacious cells for therapy; iv) inability to address the vascular compromise during SCI; v) lack of non-invasive methodologies to track the transplanted cells; vi) need for scaffolds to retain the cells at the site of injury; and vii) physical and chemical stimuli that might be required for synapses formation yielding functional neurons. EXPERT OPINION Further research on scaffolds for retaining the transplanted cells at the lesion, chemical and physical stimuli that may help neurons become functional, a meta-analysis of timing of the cell therapy, mode of application and larger clinical studies are essential to improve the outcome.
Collapse
Affiliation(s)
- Vidyasagar Devaprasad Dedeepiya
- Nichi-In Centre for Regenerative Medicine (NCRM), The Mary-Yoshio Translational Hexagon (MYTH) , PB 1262, Chennai - 600034, Tamil Nadu , India +91 44 24732186 ; ,
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Achyut BR, Varma NRS, Arbab AS. Application of Umbilical Cord Blood Derived Stem Cells in Diseases of the Nervous System. ACTA ACUST UNITED AC 2014; 4. [PMID: 25599002 DOI: 10.4172/2157-7633.1000202] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Umbilical cord blood (UCB) derived multipotent stem cells are capable of giving rise hematopoietic, epithelial, endothelial and neural progenitor cells. Thus suggested to significantly improve graft-versus-host disease and represent the distinctive therapeutic option for several malignant and non-malignant diseases. Recent advances in strategies to isolate, expand and shorten the timing of UCB stem cells engraftment have tremendously improved the efficacy of transplantations. Nervous system has limited regenerative potential in disease conditions such as cancer, neurodegeneration, stroke, and several neural injuries. This review focuses on application of UCB derived stem/progenitor cells in aforementioned pathological conditions. We have discussed the possible attempts to make use of UCB therapies to generate neural cells and tissues with developmental and functional similarities to neuronal cells. In addition, emerging applications of UCB derived AC133+ (CD133+) endothelial progenitor cells (EPCs) as imaging probe, regenerative agent, and gene delivery vehicle are mentioned that will further improve the understanding of use of UCB cells in therapeutic modalities. However, safe and effective protocols for cell transplantations are still required for therapeutic efficacy.
Collapse
Affiliation(s)
- Bhagelu R Achyut
- Tumor Angiogenesis Lab, Cancer Center, Georgia Regents University, Augusta, GA 30912, USA
| | | | - Ali S Arbab
- Tumor Angiogenesis Lab, Cancer Center, Georgia Regents University, Augusta, GA 30912, USA
| |
Collapse
|