1
|
Gordon J, Borlongan CV. An update on stem cell therapy for stroke patients: Where are we now? J Cereb Blood Flow Metab 2024; 44:1469-1479. [PMID: 38639015 PMCID: PMC11418600 DOI: 10.1177/0271678x241227022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/17/2023] [Accepted: 11/29/2023] [Indexed: 04/20/2024]
Abstract
With a foundation built upon initial work from the 1980s demonstrating graft viability in cerebral ischemia, stem cell transplantation has shown immense promise in promoting survival, enhancing neuroprotection and inducing neuroregeneration, while mitigating both histological and behavioral deficits that frequently accompany ischemic stroke. These findings have led to a number of clinical trials that have thoroughly supported a strong safety profile for stem cell therapy in patients but have generated variable efficacy. As preclinical evidence continues to expand through the investigation of new cell lines and optimization of stem cell delivery, it remains critical for translational models to adhere to the protocols established through basic scientific research. With the recent shift in approach towards utilization of stem cells as a conjunctive therapy alongside standard thrombolytic treatments, key issues including timing, route of administration, and stem cell type must each be appropriately translated from the laboratory in order to resolve the question of stem cell efficacy for cerebral ischemia that ultimately will enhance therapeutics for stroke patients towards improving quality of life.
Collapse
Affiliation(s)
- Jonah Gordon
- Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Cesar V Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| |
Collapse
|
2
|
Wu Q, Que W, Zhang J, Chen X, Wang M, Kou J, Chen G. Enhanced effect of limb remote ischemic postconditioning combined with paeoniflorin on alleviating cerebral ischemic injury via neutrophil NADPH pathway. Brain Res Bull 2024; 214:111006. [PMID: 38852654 DOI: 10.1016/j.brainresbull.2024.111006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 05/24/2024] [Accepted: 06/06/2024] [Indexed: 06/11/2024]
Abstract
BACKGROUND Limb remote ischemic postconditioning (LRIP) and paeoniflorin (PF) both can ameliorate cerebral ischemia reperfusion (I/R) injury. At present, whether LRIP combined with PF can achieve better therapeutic effect is unknown. PURPOSE This study explored the alleviating effect and mechanism of LRIP in combination with PF on cerebral I/R injury in rats. METHODS Middle cerebral artery occlusion (MCAO) surgery was performed on rats except Sham group. Then PF (2.5 mg/kg, 5 mg/kg, 10 mg/kg) was administrated by intraperitoneal injection 10 min before the start of reperfusion. LRIP was operated on the left femoral artery at 0 h of reperfusion. Behavioral testing was used to assess neurological impairment, while TTC staining was used to examine infarct volume. Protein expression of MyD88, TRAF6, p38-MAPK and phosphorylation of p47phox in neutrophils from rat peripheral blood were tested by Western blot. Rat bone marrow neutrophils were extracted and incubated for 24 h with serum from rats after LRIP combined with PF. p38 MAPK inhibitor group was administrated SB203580 while the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase inhibitor group was administrated Apocynin. Neutrophils were stimulated by fMLP (10 μM). Reactive oxygen species (ROS) production and protein expression of MyD88, TRAF6, p38 MAPK, and p47phox (ser 304 and ser 345) were detected. RESULTS LRIP combined with PF (5 mg/kg) reduced cerebral infarct volume, ameliorated neurological deficit score (NDS), decreased fMLP-stimulated ROS release and downregulated the protein expression of MyD88, TRAF6, p38-MAPK and phosphorylation of p47phox (ser 304 and ser 345) in neutrophils. CONCLUSION The protective effect of LRIP combined with PF on cerebral I/R injury was better than either alone. Taken together, we provided solid evidence to demonstrate that the combination of LRIP and PF had potential to alleviate cerebral I/R injury, which was regulated by MyD88-TRAF6-p38 MAPK pathway and neutrophil NADPH oxidase pathway.
Collapse
Affiliation(s)
- Qi Wu
- State Key Laboratory of Natural Medicines, Research Department of Pharmacognosy, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing, Jiangsu 211198, China
| | - Wenxuan Que
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing, Jiangsu 211198, China
| | - Jiangwei Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing, Jiangsu 211198, China
| | - Xun Chen
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing, Jiangsu 211198, China
| | - Min Wang
- Medical Outpatient Department, China Pharmaceutical University, 639 Longmian Road, Nanjing, Jiangsu 211198, China
| | - Junping Kou
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing, Jiangsu 211198, China.
| | - Gangling Chen
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Pharmacology of Chinese Material Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Road, Nanjing, Jiangsu 211198, China.
| |
Collapse
|
3
|
González-Burguera I, Lin G, López de Jesús M, Saumell-Esnaola M, Barrondo S, García Del Caño G, Sallés J, Scarlata S. PLCβ1 by-passes early growth response -1 to induce the differentiation of neuronal cells. Cell Death Discov 2024; 10:250. [PMID: 38789419 PMCID: PMC11126630 DOI: 10.1038/s41420-024-02009-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 04/30/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
The Gαq/phospholipase C-β (PLCβ) signaling system mediates calcium responses to a variety of hormones and neurotransmitters. Recent studies suggest that PLCβ1 expression plays a role in the differentiation of two types of cultured neuronal cells (PC12 and SK-N-SH) through a mechanism independent of Gαq. Here, we show that, similar to that observed in PC12 and SK-N-SH cells, PLCβ1 expression increases when human NT2 cells are induced to differentiate either through cytosine-β-D-arabinofuranoside or retinoic acid. Preventing this increase, abolishes differentiation, and down-regulating PLCβ1 in rat primary astrocytes causes cells to adapt an undifferentiated morphology. Surprisingly, transfecting PLCβ1 into undifferentiated PC12 or NT2 cells induces differentiation without the need for differentiating agents. Studies to uncover the underlying mechanism focused on the transcription factor early growth response 1 (Egr-1) which mediates PLCβ1 expression early in differentiation. Over-expressing PLCβ1 in HEK293 cells enhances Egr-1 expression and induces morphological changes. We show that increased levels of cytosolic PLCβ1 in undifferentiated PC12 cells disrupts the association between Egr-1 and its cytosolic binding partner (Tar RNA binding protein), promoting relocalization of Egr-1 to the nucleus, which promotes transcription of proteins needed for differentiation. These studies show a novel mechanism through which differentiation can be modulated.
Collapse
Affiliation(s)
- Imanol González-Burguera
- Department of Neurosciences, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), 01006, Vitoria-Gasteiz, Spain
- Bioaraba, Neurofarmacología Celular y Molecular, 01006, Vitoria-Gasteiz, Spain
| | - Guanyu Lin
- Department of Chemistry and Biochemistry, Worcester Polytechnic Institute, Worcester, Massachusetts, 01609, USA
| | - Maider López de Jesús
- Bioaraba, Neurofarmacología Celular y Molecular, 01006, Vitoria-Gasteiz, Spain
- Department of Pharmacology, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), 01006, Vitoria-Gasteiz, Spain
| | - Miquel Saumell-Esnaola
- Bioaraba, Neurofarmacología Celular y Molecular, 01006, Vitoria-Gasteiz, Spain
- Department of Pharmacology, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), 01006, Vitoria-Gasteiz, Spain
| | - Sergio Barrondo
- Bioaraba, Neurofarmacología Celular y Molecular, 01006, Vitoria-Gasteiz, Spain
- Department of Pharmacology, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), 01006, Vitoria-Gasteiz, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), 28029, Madrid, Spain
| | - Gontzal García Del Caño
- Department of Neurosciences, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), 01006, Vitoria-Gasteiz, Spain
- Bioaraba, Neurofarmacología Celular y Molecular, 01006, Vitoria-Gasteiz, Spain
| | - Joan Sallés
- Bioaraba, Neurofarmacología Celular y Molecular, 01006, Vitoria-Gasteiz, Spain
- Department of Pharmacology, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), 01006, Vitoria-Gasteiz, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), 28029, Madrid, Spain
| | - Suzanne Scarlata
- Department of Chemistry and Biochemistry, Worcester Polytechnic Institute, Worcester, Massachusetts, 01609, USA.
| |
Collapse
|
4
|
Andrews PW. The origins of human pluripotent stem cells: the road from a cancer to regenerative medicine. In Vitro Cell Dev Biol Anim 2024; 60:514-520. [PMID: 38396072 PMCID: PMC11126438 DOI: 10.1007/s11626-024-00865-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024]
Abstract
The notion of using pluripotent stem cells (PSCs) as a source of differentiated cell types for replacement of disease or damaged tissues in regenerative medicine is now an active area of research, with approaches to treating eye diseases such as age-related macular degeneration or Parkinson's disease now on the horizon. But the foundations for this research lie in a quite different area of science, namely the role of genetics of cancer. In this review, we trace the evolution of ideas starting with the discovery that strain 129 mice are particularly subject to develop germ cell tumors, through the identification of embryonal carcinoma (EC) cells as the stem cells of the teratocarcinoma manifestation of these tumors, to the recognition of their relationship to pluripotent cells of the early embryo, and eventually their role in the derivation of embryonic stem cells, first from mouse embryos and then from primates including humans. This is a story that illustrates how science commonly develops through the interests and insights of individual investigators, often with unexpected and unintended outcomes.
Collapse
Affiliation(s)
- Peter W Andrews
- The Centre for Stem Cell Biology, The School of Biosciences, The University of Sheffield, Western Bank, Sheffield, S10 2TN, UK.
| |
Collapse
|
5
|
Tang Y, Wang Z, Teng H, Ni H, Chen H, Lu J, Chen Z, Wang Z. Safety and efficacy of bone marrow mononuclear cell therapy for ischemic stroke recovery: a systematic review and meta-analysis of randomized controlled trials. Neurol Sci 2024; 45:1885-1896. [PMID: 38172413 PMCID: PMC11021295 DOI: 10.1007/s10072-023-07274-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 12/16/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND Cell-based therapy represents a potential treatment for ischemic stroke (IS). Here, we performed a systematic review and meta-analysis to summarize the evidence provided by randomized controlled trials (RCTs) for the transplantation of bone marrow mononuclear cells (BMMNCs) in patients with IS in any phase after stroke. METHODS We searched several databases for relevant articles up to the 10th of March 2023, including MEDLINE, EMBASE, the Cochrane Library, and ClinicalTrials.gov. Subgroup analyses were implemented to evaluate the dose and route of BMMNC administration. Statistical data were analyzed by Review Manager version 5.3 software. RESULTS Six RCTs were included in this article, including 177 patients who were treated by the transplantation of BMMNCs and 166 patients who received medical treatment. The three-month National Institutes of Health Stroke Scale (NIHSS) score indicated a favorable outcome for the BMMNC transplantation group (standardized mean difference (SMD), - 0.34; 95% confidence interval (CI), - 0.57 to - 0.11; P = 0.004). There were no significant differences between the two groups at six months post-transplantation with regards to NIHSS score (SMD 0.00; 95% CI - 0.26 to 0.27; P = 0.97), modified Rankin Scale (risk ratio (RR) 1.10; 95% CI 0.75 to 1.63; P = 0.62), Barthel Index change (SMD 0.68; 95% CI - 0.59 to 1.95; P = 0.29), and infarct volume change (SMD - 0.08; 95% CI - 0.42 to 0.26; P = 0.64). In addition, there was no significant difference between the two groups in terms of safety outcome (RR 1.24; 95% CI 0.80 to 1.91; P = 0.33). CONCLUSION Our meta-analysis demonstrated that the transplantation of BMMNCs was safe; however, the efficacy of this procedure requires further validation in larger RTCs.
Collapse
Affiliation(s)
- Yanbing Tang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China
- Suzhou Medical College of Soochow University, Suzhou, 215002, Jiangsu Province, China
| | - Zilan Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China
| | - Haiying Teng
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China
| | - Hanyu Ni
- Suzhou Medical College of Soochow University, Suzhou, 215002, Jiangsu Province, China
| | - Huiru Chen
- Department of Neurology, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China
| | - Jiaye Lu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China
| | - Zhouqing Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China.
| | - Zhong Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China.
| |
Collapse
|
6
|
Sanberg PR, Morrison D, Bjugstad KB. 30 years of American Society for Neural Therapy and Repair (ASNTR): A Personal Perspective at the Intersection of Science, Politics, and Culture. Neurosci Biobehav Rev 2023; 151:105234. [PMID: 37196924 DOI: 10.1016/j.neubiorev.2023.105234] [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: 04/11/2023] [Revised: 05/09/2023] [Accepted: 05/14/2023] [Indexed: 05/19/2023]
Abstract
The American Society for Neural Therapy and Repair (ASNTR) started 30 years ago in 1993 as the American Society for Neural Transplantation (ASNT), with an emphasis on neural transplantation. Through the years, the Society has been shaped as much by our expanding knowledge of neurodegenerative disorders and how to treat them as it has by politics and culture. What once felt like a leash on neuroscience research, has turned into an advantage as neural transplantation evolved into neural therapy and repair. This brief commentary provides a personalized account of our research during the Society's years.
Collapse
Affiliation(s)
- Paul R Sanberg
- Center of Excellence for Aging and Brain Repair, Depts of Neurosurgery and Brain Repair, Pathology and Cell Biology, and Psychiatry. University of South Florida, Morsani College of Medicine, Tampa, FL 33612.
| | - Donna Morrison
- Center of Excellence for Aging and Brain Repair, Depts of Neurosurgery and Brain Repair, Pathology and Cell Biology, and Psychiatry. University of South Florida, Morsani College of Medicine, Tampa, FL 33612
| | - Kimberly B Bjugstad
- Center of Excellence for Aging and Brain Repair, Depts of Neurosurgery and Brain Repair, Pathology and Cell Biology, and Psychiatry. University of South Florida, Morsani College of Medicine, Tampa, FL 33612
| |
Collapse
|
7
|
Bjugstad K, Sanberg P. The boundlessness of behavioral neuroscience: A look across 30 years. Neurosci Biobehav Rev 2022; 142:104910. [DOI: 10.1016/j.neubiorev.2022.104910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 10/06/2022] [Indexed: 11/30/2022]
|
8
|
Abstract
The last 50 years have witnessed the translation of stem cell therapy from the laboratory to the clinic for treating brain disorders, in particular stroke. From the focal stereotaxic transplantation to the minimally invasive intravenous and intraarterial delivery, stem cells display the ability to replenish injured cells and to secrete therapeutic molecules, altogether promoting brain repair. The increased stroke incidence in COVID-19 survivors poses as a new disease indication for cell therapy, owing in part to the cells' robust anti-inflammatory properties. Optimization of the cell transplant regimen will ensure the safe and effective clinical application of cell therapy in stroke and relevant neurological disorders.
Collapse
Affiliation(s)
- You Jeong Park
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, USA
| | - Cesar V Borlongan
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, USA
| |
Collapse
|
9
|
Gu BJ, Kung DK, Chen HCI. Cell Therapy for Stroke: A Mechanistic Analysis. Neurosurgery 2021; 88:733-745. [PMID: 33370810 DOI: 10.1093/neuros/nyaa531] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 09/26/2020] [Indexed: 11/12/2022] Open
Abstract
Cell therapy has been widely recognized as a promising strategy to enhance recovery in stroke survivors. However, despite an abundance of encouraging preclinical data, successful clinical translation remains elusive. As the field continues to advance, it is important to reexamine prior clinical trials in the context of their intended mechanisms, as this can inform future preclinical and translational efforts. In the present work, we review the major clinical trials of cell therapy for stroke and highlight a mechanistic shift between the earliest studies, which aimed to replace dead and damaged neurons, and later ones that focused on exploiting the various neuromodulatory effects afforded by stem cells. We discuss why both mechanisms are worth pursuing and emphasize the means through which cell replacement can still be achieved.
Collapse
Affiliation(s)
- Ben Jiahe Gu
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - David K Kung
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Han-Chiao Isaac Chen
- Department of Neurosurgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
10
|
Abstract
The role of cellular transplantation to promote functional recovery after stroke has been evaluated over the last two decades. Preclinical studies first established the potential for cultured neuronal cells derived from a teratocarcinoma cell line to be tested for safety and efficacy in the treatment of human stroke. In animal models of stroke that caused reproducible learning and motor deficits, injection of neuronal cells resulted in a return of learning behavior, retention time, and motor function. Clinical trials followed. Additional work with cells derived from a bone marrow neuroprogenitor line, fetal cortical stem cells, and other cell sources showed promise in preclinical studies and then these cells were tested in clinical studies. This report reviews the different biological repair approaches using cell implants, discusses clinical trial design and surgical methods, and the current state of research.
Collapse
Affiliation(s)
- Douglas Kondziolka
- Department of Neurosurgery, NYU Langone Health, New York University, New York, NY
| |
Collapse
|
11
|
Jiao Y, Liu YW, Chen WG, Liu J. Neuroregeneration and functional recovery after stroke: advancing neural stem cell therapy toward clinical application. Neural Regen Res 2021; 16:80-92. [PMID: 32788451 PMCID: PMC7818886 DOI: 10.4103/1673-5374.286955] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Stroke is a main cause of death and disability worldwide. The ability of the brain to self-repair in the acute and chronic phases after stroke is minimal; however, promising stem cell-based interventions are emerging that may give substantial and possibly complete recovery of brain function after stroke. Many animal models and clinical trials have demonstrated that neural stem cells (NSCs) in the central nervous system can orchestrate neurological repair through nerve regeneration, neuron polarization, axon pruning, neurite outgrowth, repair of myelin, and remodeling of the microenvironment and brain networks. Compared with other types of stem cells, NSCs have unique advantages in cell replacement, paracrine action, inflammatory regulation and neuroprotection. Our review summarizes NSC origins, characteristics, therapeutic mechanisms and repair processes, then highlights current research findings and clinical evidence for NSC therapy. These results may be helpful to inform the direction of future stroke research and to guide clinical decision-making.
Collapse
Affiliation(s)
- Yang Jiao
- Stem Cell Clinical Research Center, National Joint Engineering Laboratory, Regenerative Medicine Center, The First Affiliated Hospital of Dalian Medical University; Dalian Innovation Institute of Stem Cells and Precision Medicine, Dalian, Liaoning Province, China
| | - Yu-Wan Liu
- Stem Cell Clinical Research Center, National Joint Engineering Laboratory, Regenerative Medicine Center, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Wei-Gong Chen
- Stem Cell Clinical Research Center, National Joint Engineering Laboratory, Regenerative Medicine Center, The First Affiliated Hospital of Dalian Medical University; Dalian Innovation Institute of Stem Cells and Precision Medicine, Dalian, Liaoning Province, China
| | - Jing Liu
- Stem Cell Clinical Research Center, National Joint Engineering Laboratory, Regenerative Medicine Center, The First Affiliated Hospital of Dalian Medical University; Dalian Innovation Institute of Stem Cells and Precision Medicine, Dalian, Liaoning Province, China
| |
Collapse
|
12
|
He JQ, Sussman ES, Steinberg GK. Revisiting Stem Cell-Based Clinical Trials for Ischemic Stroke. Front Aging Neurosci 2020; 12:575990. [PMID: 33381020 PMCID: PMC7767918 DOI: 10.3389/fnagi.2020.575990] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 11/23/2020] [Indexed: 12/12/2022] Open
Abstract
Stroke is the leading cause of serious long-term disability, significantly reducing mobility in almost half of the affected patients aged 65 years and older. There are currently no proven neurorestorative treatments for chronic stroke. To address the complex problem of restoring function in ischemic brain tissue, stem cell transplantation-based therapies have emerged as potential restorative therapies. Aligning with the major cell types found within the ischemic brain, stem-cell-based clinical trials for ischemic stroke have fallen under three broad cell lineages: hematopoietic, mesenchymal, and neural. In this review article, we will discuss the scientific rationale for transplanting cells from each of these lineages and provide an overview of published and ongoing trials using this framework.
Collapse
Affiliation(s)
- Joy Q He
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Eric S Sussman
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Gary K Steinberg
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, United States.,Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, United States.,Stanford Stroke Center, Stanford Health Care, Stanford, CA, United States
| |
Collapse
|
13
|
Egawa N, Suzuki H, Takahashi R, Hayakawa K, Li W, Lo EH, Arai K, Inoue H. From in vitro to in vivo reprogramming for neural transdifferentiation: An approach for CNS tissue remodeling using stem cell technology. J Cereb Blood Flow Metab 2020; 40:1739-1751. [PMID: 32423328 PMCID: PMC7446571 DOI: 10.1177/0271678x20910324] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Advances in stem cell technology have provided three approaches to address the demanding issue of the treatment of intractable neurological disease. One of the approaches is the screening of compounds attenuating pathological phenotypes in stem-cell based models. A second approach consists of exogenous-targeted cell supplementation to the lesion with stem cell-derived differentiated cells. A third approach involves in vivo direct programming to transdifferentiate endogenous somatic cells and to boost CNS tissue remodeling. In this review, we outline research advances in stem cell technology of direct reprogramming in vitro and in vivo and discuss the future challenge of tissue remodeling by neural transdifferentiation.
Collapse
Affiliation(s)
- Naohiro Egawa
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan.,iPSC-based Drug Discovery and Development Team, RIKEN BioResource Research Center (BRC), Kyoto, Japan.,Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Hidefumi Suzuki
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan.,iPSC-based Drug Discovery and Development Team, RIKEN BioResource Research Center (BRC), Kyoto, Japan.,Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Ryosuke Takahashi
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kazuhide Hayakawa
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Wenlu Li
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Eng H Lo
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ken Arai
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Haruhisa Inoue
- iPSC-based Drug Discovery and Development Team, RIKEN BioResource Research Center (BRC), Kyoto, Japan.,Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan.,Medical-risk Avoidance based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP), Kyoto, Japan
| |
Collapse
|
14
|
Mangin G, Kubis N. Cell Therapy for Ischemic Stroke: How to Turn a Promising Preclinical Research into a Successful Clinical Story. Stem Cell Rev Rep 2020; 15:176-193. [PMID: 30443706 DOI: 10.1007/s12015-018-9864-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Stroke is a major public health issue with limited treatment. The pharmacologically or mechanically removing of the clot is accessible to less than 10% of the patients. Stem cell therapy is a promising alternative strategy since it increases the therapeutic time window but many issues remain unsolved. To avoid a new dramatic failure when translating experimental data on the bedside, this review aims to highlight the indispensable checkpoints to make a successful clinical trial based on the current preclinical literature. The large panel of progenitors/ stem cells at the researcher's disposal is to be used wisely, regarding the type of cells, the source of cells, the route of delivery, the time window, since it will directly affect the outcome. Mechanisms are still incompletely understood, although recent studies have focused on the inflammation modulation of most cells types.
Collapse
Affiliation(s)
| | - Nathalie Kubis
- INSERM U965, F-75475, Paris, France. .,Sorbonne Paris Cité, Université Paris Diderot, F-75475, Paris, France. .,Service de Physiologie Clinique-Explorations Fonctionnelles, AP-HP, Hôpital Lariboisière, 2 rue Ambroise Paré, F-75475, Paris, France.
| |
Collapse
|
15
|
Corey S, Bonsack B, Heyck M, Shear A, Sadanandan N, Zhang H, Borlongan CV. Harnessing the anti-inflammatory properties of stem cells for transplant therapy in hemorrhagic stroke. BRAIN HEMORRHAGES 2020; 1:24-33. [PMID: 34056567 PMCID: PMC8158660 DOI: 10.1016/j.hest.2019.12.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Hemorrhagic stroke is a global health crisis plagued by neuroinflammation in the acute and chronic phases. Neuroinflammation approximates secondary cell death, which in turn robustly contributes to stroke pathology. Both the physiological and behavioral symptoms of stroke correlate with various inflammatory responses in animal and human studies. That slowing the secondary cell death mediated by this inflammation may attenuate stroke pathology presents a novel treatment strategy. To this end, experimental therapies employing stem cell transplants support their potential for neuroprotection and neuroregeneration after hemorrhagic stroke. In this review, we evaluate experiments using different types of stem cell transplants as treatments for stroke-induced neuroinflammation. We also update this emerging area by examining recent preclinical and clinical trials that have deployed these therapies. While further investigations are warranted to solidify their therapeutic profile, the reviewed studies largely posit stem cells as safe and potent biologics for stroke, specifically owing to their mode of action for sequestering neuroinflammation and promoting neuroregenerative processes.
Collapse
Affiliation(s)
- Sydney Corey
- Center of Excellence for Aging and Brain Repair, University of South Florida, College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| | - Brooke Bonsack
- Center of Excellence for Aging and Brain Repair, University of South Florida, College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| | - Matt Heyck
- Center of Excellence for Aging and Brain Repair, University of South Florida, College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| | - Alex Shear
- Center of Excellence for Aging and Brain Repair, University of South Florida, College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| | - Nadia Sadanandan
- Center of Excellence for Aging and Brain Repair, University of South Florida, College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| | - Henry Zhang
- Center of Excellence for Aging and Brain Repair, University of South Florida, College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| | - Cesar V Borlongan
- Center of Excellence for Aging and Brain Repair, University of South Florida, College of Medicine, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| |
Collapse
|
16
|
Krucoff MO, Miller JP, Saxena T, Bellamkonda R, Rahimpour S, Harward SC, Lad SP, Turner DA. Toward Functional Restoration of the Central Nervous System: A Review of Translational Neuroscience Principles. Neurosurgery 2020; 84:30-40. [PMID: 29800461 DOI: 10.1093/neuros/nyy128] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 03/15/2018] [Indexed: 01/09/2023] Open
Abstract
Injury to the central nervous system (CNS) can leave patients with devastating neurological deficits that may permanently impair independence and diminish quality of life. Recent insights into how the CNS responds to injury and reacts to critically timed interventions are being translated into clinical applications that have the capacity to drastically improve outcomes for patients suffering from permanent neurological deficits due to spinal cord injury, stroke, or other CNS disorders. The translation of such knowledge into practical and impactful treatments involves the strategic collaboration between neurosurgeons, clinicians, therapists, scientists, and industry. Therefore, a common understanding of key neuroscientific principles is crucial. Conceptually, current approaches to CNS revitalization can be divided by scale into macroscopic (systems-circuitry) and microscopic (cellular-molecular). Here we review both emerging and well-established tenets that are being utilized to enhance CNS recovery on both levels, and we explore the role of neurosurgeons in developing therapies moving forward. Key principles include plasticity-driven functional recovery, cellular signaling mechanisms in axonal sprouting, critical timing for recovery after injury, and mechanisms of action underlying cellular replacement strategies. We then discuss integrative approaches aimed at synergizing interventions across scales, and we make recommendations for the basis of future clinical trial design. Ultimately, we argue that strategic modulation of microscopic cellular behavior within a macroscopic framework of functional circuitry re-establishment should provide the foundation for most neural restoration strategies, and the early involvement of neurosurgeons in the process will be crucial to successful clinical translation.
Collapse
Affiliation(s)
- Max O Krucoff
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
| | - Jonathan P Miller
- Department of Neurosurgery, Case Western Reserve University, Cleve-land, Ohio
| | - Tarun Saxena
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Ravi Bellamkonda
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Shervin Rahimpour
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
| | - Stephen C Harward
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
| | - Shivanand P Lad
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina.,Department of Mechan-ical Engineering and Material Sciences, Pratt School of Engineering, Duke Uni-versity, Durham, North Carolina.,Duke Institute for Brain Sciences, Duke Univer-sity, Durham, North Carolina.,Research and Surgery Services, Durham Veterans Affairs Medical Center, Durham, North Carolina
| | - Dennis A Turner
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina.,Department of Biomedical Engineering, Duke University, Durham, North Carolina.,Depart-ment of Neurobiology, Duke University, Durham, North Carolina.,Research and Surgery Services, Durham Veterans Affairs Medical Center, Durham, North Carolina
| |
Collapse
|
17
|
Wang Y, Graham ES, Unsworth CP. First observations of spontaneous bursting in human hNT neurons with a customised neural chip platform. Biomed Phys Eng Express 2019. [DOI: 10.1088/2057-1976/ab4b24] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
18
|
Tuazon JP, Castelli V, Borlongan CV. Drug-like delivery methods of stem cells as biologics for stroke. Expert Opin Drug Deliv 2019; 16:823-833. [PMID: 31311344 DOI: 10.1080/17425247.2019.1645116] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Introduction: Stem cell therapy is an experimental treatment for brain disorders. Although a cellular product, stem cells can be classified as biologics based on the cells' secretion of therapeutic substances. Treatment with stem cell biologics may appeal to stroke because of the secondary cell death mechanisms, especially neuroinflammation, that are rampant from the onset and remain elevated during the progressive phase of the disease requiring multi-pronged biological targets to effectively abrogate the neurodegenerative pathology. However, the optimal delivery methods, among other logistical approaches (i.e. cell doses and timing of intervention), for stem cell therapy will need to be refined before stem cell biologics can be successfully utilized for stroke in large scale clinical trials. Areas covered: In this review, we discuss how the innate qualities of stem cells characterize them as biologics, how stem cell transplantation may be an ideal treatment for stroke, and the various routes of stem cell administration that have been employed in various preclinical and clinical investigations. Expert opinion: There is a need to optimize the delivery of stem cell biologics for stroke in order to guide the safe and effective translation of this therapy from the laboratory to the clinic.
Collapse
Affiliation(s)
- Julian P Tuazon
- a Center of Excellence for Aging & Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine , Tampa , FL , USA
| | - Vanessa Castelli
- a Center of Excellence for Aging & Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine , Tampa , FL , USA
| | - Cesar V Borlongan
- a Center of Excellence for Aging & Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine , Tampa , FL , USA
| |
Collapse
|
19
|
Borlongan CV. Concise Review: Stem Cell Therapy for Stroke Patients: Are We There Yet? Stem Cells Transl Med 2019; 8:983-988. [PMID: 31099181 PMCID: PMC6708064 DOI: 10.1002/sctm.19-0076] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 05/03/2019] [Indexed: 12/14/2022] Open
Abstract
Four decades of preclinical research demonstrating survival, functional integration, and behavioral effects of transplanted stem cells in experimental stroke models have provided ample scientific basis for initiating limited clinical trials of stem cell therapy in stroke patients. Although safety of the grafted cells has been overwhelmingly documented, efficacy has not been forthcoming. Two recently concluded stroke clinical trials on mesenchymal stem cells (MSCs) highlight the importance of strict adherence to the basic science findings of optimal transplant regimen of cell dose, timing, and route of delivery in enhancing the functional outcomes of cell therapy. Echoing the Stem Cell Therapeutics as an Emerging Paradigm for Stroke and Stroke Treatment Academic Industry Roundtable call for an NIH‐guided collaborative consortium of multiple laboratories in testing the safety and efficacy of stem cells and their derivatives, not just as stand‐alone but preferably in combination with approved thrombolytic or thrombectomy, may further increase the likelihood of successful fruition of translating stem cell therapy for stroke clinical application. The laboratory and clinical experience with MSC therapy for stroke may guide the future translational research on stem cell‐based regenerative medicine in neurological disorders. stem cells translational medicine2019;8:983&988
Collapse
Affiliation(s)
- Cesario V Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, Florida, USA
| |
Collapse
|
20
|
Schwarz S, Spitzbarth I, Baumgärtner W, Lehmbecker A. Cryopreservation of Canine Primary Dorsal Root Ganglion Neurons and Its Impact upon Susceptibility to Paramyxovirus Infection. Int J Mol Sci 2019; 20:ijms20051058. [PMID: 30823498 PMCID: PMC6429404 DOI: 10.3390/ijms20051058] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 02/22/2019] [Accepted: 02/25/2019] [Indexed: 12/26/2022] Open
Abstract
Canine dorsal root ganglion (DRG) neurons, isolated post mortem from adult dogs, could provide a promising tool to study neuropathogenesis of neurotropic virus infections with a non-rodent host spectrum. However, access to canine DRG is limited due to lack of donor tissue and the cryopreservation of DRG neurons would greatly facilitate experiments. The present study aimed (i) to establish canine DRG neurons as an in vitro model for canine distemper virus (CDV) infection; and (ii) to determine whether DRG neurons are cryopreservable and remain infectable with CDV. Neurons were characterized morphologically and phenotypically by light microscopy, immunofluorescence, and functionally, by studying their neurite outgrowth and infectability with CDV. Cryopreserved canine DRG neurons remained in culture for at least 12 days. Furthermore, both non-cryopreserved and cryopreserved DRG neurons were susceptible to infection with two different strains of CDV, albeit only one of the two strains (CDV R252) provided sufficient absolute numbers of infected neurons. However, cryopreserved DRG neurons showed reduced cell yield, neurite outgrowth, neurite branching, and soma size and reduced susceptibility to CDV infection. In conclusion, canine primary DRG neurons represent a suitable tool for investigations upon the pathogenesis of neuronal CDV infection. Moreover, despite certain limitations, cryopreserved canine DRG neurons generally provide a useful and practicable alternative to address questions regarding virus tropism and neuropathogenesis.
Collapse
Affiliation(s)
- Sarah Schwarz
- Department of Pathology, University of Veterinary Medicine, 30559 Hannover, Germany.
- Center for Systems Neuroscience, 30559 Hannover, Germany.
| | - Ingo Spitzbarth
- Department of Pathology, University of Veterinary Medicine, 30559 Hannover, Germany.
- Center for Systems Neuroscience, 30559 Hannover, Germany.
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine, 30559 Hannover, Germany.
- Center for Systems Neuroscience, 30559 Hannover, Germany.
| | - Annika Lehmbecker
- Department of Pathology, University of Veterinary Medicine, 30559 Hannover, Germany.
- Center for Systems Neuroscience, 30559 Hannover, Germany.
| |
Collapse
|
21
|
Lee JY, Acosta S, Tuazon JP, Xu K, Nguyen H, Lippert T, Liska MG, Semechkin A, Garitaonandia I, Gonzalez R, Kern R, Borlongan CV. Human parthenogenetic neural stem cell grafts promote multiple regenerative processes in a traumatic brain injury model. Am J Cancer Res 2019; 9:1029-1046. [PMID: 30867814 PMCID: PMC6401413 DOI: 10.7150/thno.29868] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 11/16/2018] [Indexed: 12/12/2022] Open
Abstract
International Stem Cell Corporation human parthenogenetic neural stem cells (ISC-hpNSC) have potential therapeutic value for patients suffering from traumatic brain injury (TBI). Here, we demonstrate the behavioral and histological effects of transplanting ISC-hpNSC intracerebrally in an animal model of TBI. Methods: Sprague-Dawley rats underwent a moderate controlled cortical impact TBI surgery. Transplantation occurred at 72 h post-TBI with functional readouts of behavioral and histological deficits conducted during the subsequent 3-month period after TBI. We characterized locomotor, neurological, and cognitive performance at baseline (before TBI), then on days 0, 1, 7, 14, 30, 60, and 90 (locomotor and neurological), and on days 28-30, 58-60, and 88-90 (cognitive) after TBI. Following completion of behavioral testing at 3 months post-TBI, animals were euthanized by transcardial perfusion and brains harvested to histologically characterize the extent of brain damage. Neuronal survival was revealed by Nissl staining, and stem cell engraftment and host tissue repair mechanisms such as the anti-inflammatory response in peri-TBI lesion areas were examined by immunohistochemical analyses. Results: We observed that TBI groups given high and moderate doses of ISC-hpNSC had an improved swing bias on an elevated body swing test for motor function, increased scores on forelimb akinesia and paw grasp neurological tests, and committed significantly fewer errors on a radial arm water maze test for cognition. Furthermore, histological analyses indicated that high and moderate doses of stem cells increased the expression of phenotypic markers related to the neural lineage and myelination and decreased reactive gliosis and inflammation in the brain, increased neuronal survival in the peri-impact area of the cortex, and decreased inflammation in the spleen at 90 days post-TBI. Conclusion: These results provide evidence that high and moderate doses of ISC-hpNSC ameliorate TBI-associated histological alterations and motor, neurological, and cognitive deficits.
Collapse
|
22
|
González-Nieto D, Fernández-García L, Pérez-Rigueiro J, Guinea GV, Panetsos F. Hydrogels-Assisted Cell Engraftment for Repairing the Stroke-Damaged Brain: Chimera or Reality. Polymers (Basel) 2018; 10:polym10020184. [PMID: 30966220 PMCID: PMC6415003 DOI: 10.3390/polym10020184] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 02/06/2018] [Accepted: 02/11/2018] [Indexed: 01/07/2023] Open
Abstract
The use of advanced biomaterials as a structural and functional support for stem cells-based therapeutic implants has boosted the development of tissue engineering applications in multiple clinical fields. In relation to neurological disorders, we are still far from the clinical reality of restoring normal brain function in neurodegenerative diseases and cerebrovascular disorders. Hydrogel polymers show unique mechanical stiffness properties in the range of living soft tissues such as nervous tissue. Furthermore, the use of these polymers drastically enhances the engraftment of stem cells as well as their capacity to produce and deliver neuroprotective and neuroregenerative factors in the host tissue. Along this article, we review past and current trends in experimental and translational research to understand the opportunities, benefits, and types of tentative hydrogel-based applications for the treatment of cerebral disorders. Although the use of hydrogels for brain disorders has been restricted to the experimental area, the current level of knowledge anticipates an intense development of this field to reach clinics in forthcoming years.
Collapse
Affiliation(s)
- Daniel González-Nieto
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28040 Madrid, Spain.
- Departamento de Tecnología Fotónica y Bioingeniería, ETSI Telecomunicaciones, Universidad Politécnica de Madrid, 28040 Madrid, Spain.
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), 28040 Madrid, Spain.
| | - Laura Fernández-García
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28040 Madrid, Spain.
| | - José Pérez-Rigueiro
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28040 Madrid, Spain.
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), 28040 Madrid, Spain.
- Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid 28040 Madrid, Spain.
| | - Gustavo V Guinea
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28040 Madrid, Spain.
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), 28040 Madrid, Spain.
- Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid 28040 Madrid, Spain.
| | - Fivos Panetsos
- Neurocomputing and Neurorobotics Research Group: Faculty of Biology and Faculty of Optics, Universidad Complutense de Madrid, 28040 Madrid, Spain.
- Instituto de Investigación Sanitaria, Hospital Clínico San Carlos Madrid, IdISSC, 28040 Madrid, Spain.
| |
Collapse
|
23
|
Kotoda M, Ishiyama T, Mitsui K, Hishiyama S, Matsukawa T. Neuroprotective effects of amiodarone in a mouse model of ischemic stroke. BMC Anesthesiol 2017; 17:168. [PMID: 29216818 PMCID: PMC5721470 DOI: 10.1186/s12871-017-0459-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 11/27/2017] [Indexed: 11/24/2022] Open
Abstract
Background Ion channels play a crucial role in the development of ischemic brain injury. Recent studies have reported that the blockade of various types of ion channels improves outcomes in experimental stroke models. Amiodarone, one of the most effective drugs for life-threatening arrhythmia, works as a multiple channel blocker and its characteristics cover all four Vaughan-Williams classes. Although it is known that amiodarone indirectly contributes to preventing ischemic stroke by maintaining sinus rhythm in patients with atrial fibrillation, the direct neuroprotective effect of amiodarone has not been clarified. The purpose of this study was to investigate the direct effect of amiodarone on ischemic stroke in mice. Methods Focal cerebral ischemia was induced via distal permanent middle cerebral artery occlusion (MCAO) in adult male mice. The amiodarone pre-treatment group received 50 mg/kg of amiodarone 1 h before MCAO; the amiodarone post-treatment groups received 50 mg/kg of amiodarone immediately after MCAO; the control group received vehicle only. In addition, the sodium channel opener veratrine and selective beta-adrenergic agonist isoprotelenol were used to elucidate the targeted pathway. Heart rate and blood pressure were monitored perioperatively. Infarct volume analysis was conducted 48 h after MCAO. The body asymmetry test and the corner test were used for neurological evaluation. Results Amiodarone pre-treatment and post-treatment reduced the heart rate but did not affect the blood pressure. No mice showed arrhythmia. Compared with the control group, the amiodarone pre-treatment group had smaller infarct volumes (8.9 ± 2.1% hemisphere [mean ± SD] vs. 11.2 ± 1.4%; P < 0.05) and improved functional outcomes: lower asymmetric body swing rates (52 ± 17% vs. 65 ± 18%; P < 0.05) and fewer left turns (7.1 ± 1.2 vs. 8.3 ± 1.2; P < 0.05). In contrast, amiodarone post-treatment did not improve the outcomes after MCAO. The neuroprotective effect of amiodarone pre-treatment was abolished by co-administration of veratrine but not by isoproterenol. Conclusions Amiodarone pre-treatment attenuated ischemic brain injury and improved functional outcomes without affecting heart rhythm and blood pressure. The present results showed that amiodarone pre-treatment has neuroprotective effects, at least in part, via blocking the sodium channels.
Collapse
Affiliation(s)
- Masakazu Kotoda
- Department of Anesthesiology, Faculty of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409-3898, Japan.
| | - Tadahiko Ishiyama
- Surgical Center, University of Yamanashi Hospital, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409-3898, Japan
| | - Kazuha Mitsui
- Department of Anesthesiology, Faculty of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409-3898, Japan
| | - Sohei Hishiyama
- Department of Anesthesiology, Faculty of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409-3898, Japan
| | - Takashi Matsukawa
- Department of Anesthesiology, Faculty of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 409-3898, Japan
| |
Collapse
|
24
|
Acosta S, Tran T, Mair J, Okonkwo O, Larose B, Borlongan CV. Media coverage and public awareness on bioethics perception of emerging biomedical therapies. Chin Neurosurg J 2017. [DOI: 10.1186/s41016-016-0062-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
25
|
Chang CF, Lai JH, Wu JCC, Greig NH, Becker RE, Luo Y, Chen YH, Kang SJ, Chiang YH, Chen KY. (-)-Phenserine inhibits neuronal apoptosis following ischemia/reperfusion injury. Brain Res 2017; 1677:118-128. [PMID: 28963051 DOI: 10.1016/j.brainres.2017.09.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 09/11/2017] [Accepted: 09/12/2017] [Indexed: 12/21/2022]
Abstract
Stroke commonly leads to adult disability and death worldwide. Its major symptoms are spastic hemiplegia and discordant motion, consequent to neuronal cell death induced by brain vessel occlusion. Acetylcholinesterase (AChE) is upregulated and allied with inflammation and apoptosis after stroke. Recent studies suggest that AChE inhibition ameliorates ischemia-reperfusion injury and has neuroprotective properties. (-)-Phenserine, a reversible AChE inhibitor, has a broad range of actions independent of its AChE properties, including neuroprotective ones. However, its protective effects and detailed mechanism of action in the rat middle cerebral artery occlusion model (MCAO) remain to be elucidated. This study investigated the therapeutic effects of (-)-phenserine for stroke in the rat focal cerebral ischemia model and oxygen-glucose deprivation/reperfusion (OGD/RP) damage model in SH-SY5Y neuronal cultures. (-)-Phenserine mitigated OGD/PR-induced SH-SY5Y cell death, providing an inverted U-shaped dose-response relationship between concentration and survival. In MCAO challenged rats, (-)-phenserine reduced infarction volume, cell death and improved body asymmetry, a behavioral measure of stoke impact. In both cellular and animal studies, (-)-phenserine elevated brain-derived neurotrophic factor (BDNF) and B-cell lymphoma 2 (Bcl-2) levels, and decreased activated-caspase 3, amyloid precursor protein (APP) and glial fibrillary acidic protein (GFAP) expression, potentially mediated through the ERK-1/2 signaling pathway. These actions mitigated neuronal apoptosis in the stroke penumbra, and decreased matrix metallopeptidase-9 (MMP-9) expression. In synopsis, (-)-phenserine significantly reduced neuronal damage induced by ischemia/reperfusion injury in a rat model of MCAO and cellular model of OGD/RP, demonstrating that its anti-apoptotic/neuroprotective/neurotrophic cholinergic and non-cholinergic properties warrant further evaluation in conditions of brain injury.
Collapse
Affiliation(s)
- Cheng-Fu Chang
- Department of Neurosurgery, Taipei City Hospital, Zhongxiao Branch, Taiwan; Department of Surgery, College of Medicine, Taipei Medical University, Taipei, Taiwan; Translational Laboratory, Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan
| | - Jing-Huei Lai
- Department of Surgery, College of Medicine, Taipei Medical University, Taipei, Taiwan; Translational Laboratory, Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan; Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei, Taiwan
| | - John Chung-Che Wu
- Department of Surgery, College of Medicine, Taipei Medical University, Taipei, Taiwan; Translational Laboratory, Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan; Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei, Taiwan; Department of Neurosurgery, Taipei Medical University Hospital, Taipei, Taiwan
| | - Nigel H Greig
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA.
| | - Robert E Becker
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA; Aristea Translational Medicine, Park City, UT, USA
| | - Yu Luo
- Department of Neurosurgery, Case Western Reserve University, School of Medicine, Cleveland, OH, USA
| | - Yen-Hua Chen
- Department of Surgery, College of Medicine, Taipei Medical University, Taipei, Taiwan; Translational Laboratory, Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan; Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei, Taiwan
| | - Shuo-Jhen Kang
- Department of Surgery, College of Medicine, Taipei Medical University, Taipei, Taiwan; Translational Laboratory, Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan; Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei, Taiwan
| | - Yung-Hsiao Chiang
- Department of Surgery, College of Medicine, Taipei Medical University, Taipei, Taiwan; Translational Laboratory, Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan; Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei, Taiwan; Department of Neurosurgery, Taipei Medical University Hospital, Taipei, Taiwan; Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.
| | - Kai-Yun Chen
- Translational Laboratory, Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan; Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
26
|
Stem cell therapy for abrogating stroke-induced neuroinflammation and relevant secondary cell death mechanisms. Prog Neurobiol 2017; 158:94-131. [PMID: 28743464 DOI: 10.1016/j.pneurobio.2017.07.004] [Citation(s) in RCA: 171] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Revised: 07/18/2017] [Accepted: 07/18/2017] [Indexed: 12/13/2022]
Abstract
Ischemic stroke is a leading cause of death worldwide. A key secondary cell death mechanism mediating neurological damage following the initial episode of ischemic stroke is the upregulation of endogenous neuroinflammatory processes to levels that destroy hypoxic tissue local to the area of insult, induce apoptosis, and initiate a feedback loop of inflammatory cascades that can expand the region of damage. Stem cell therapy has emerged as an experimental treatment for stroke, and accumulating evidence supports the therapeutic efficacy of stem cells to abrogate stroke-induced inflammation. In this review, we investigate clinically relevant stem cell types, such as hematopoietic stem cells (HSCs), mesenchymal stem cells (MSCs), endothelial progenitor cells (EPCs), very small embryonic-like stem cells (VSELs), neural stem cells (NSCs), extraembryonic stem cells, adipose tissue-derived stem cells, breast milk-derived stem cells, menstrual blood-derived stem cells, dental tissue-derived stem cells, induced pluripotent stem cells (iPSCs), teratocarcinoma-derived Ntera2/D1 neuron-like cells (NT2N), c-mycER(TAM) modified NSCs (CTX0E03), and notch-transfected mesenchymal stromal cells (SB623), comparing their potential efficacy to sequester stroke-induced neuroinflammation and their feasibility as translational clinical cell sources. To this end, we highlight that MSCs, with a proven track record of safety and efficacy as a transplantable cell for hematologic diseases, stand as an attractive cell type that confers superior anti-inflammatory effects in stroke both in vitro and in vivo. That stem cells can mount a robust anti-inflammatory action against stroke complements the regenerative processes of cell replacement and neurotrophic factor secretion conventionally ascribed to cell-based therapy in neurological disorders.
Collapse
|
27
|
Stewart R, Lako M, Horrocks GM, Przyborski SA. Neural Development by Transplanted Human Embryonal Carcinoma Stem Cells Expressing Green Fluorescent Protein. Cell Transplant 2017; 14:339-51. [PMID: 16180653 DOI: 10.3727/000000005783982945] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
For many years, researchers have investigated the fate and potential of neuroectodermal cells during the development of the central nervous system. Although several key factors that regulate neural differentiation have been identified, much remains unknown about the molecular mechanisms that control the fate and specification of neural subtypes, especially in humans. Human embryonal carcinoma (EC) stem cells are valuable research tools for the study of neural development; however, existing in vitro experiments are limited to inducing the differentiation of EC cells into only a handful of cell types. In this study, we developed and characterized a novel EC cell line (termed TERA2.cl.SP12-GFP) that carries the reporter molecule, green fluorescent protein (GFP). We demonstrate that TERA2.cl.SP12-GFP stem cells and their differentiated neural derivatives constitutively express GFP in cells grown both in vitro and in vivo. Cellular differentiation does not appear to be affected by insertion of the transgene. We propose that TERA2.cl.SP12-GFP cells provide a valuable research tool to track the fate of cells subsequent to transplantation into alternative environments and that this approach may be particularly useful to investigate the differentiation of human neural tissues in response to local environmental signals.
Collapse
Affiliation(s)
- R Stewart
- School of Biological and Biomedical Science, University of Durham, South Road, Durham DH1 3LE, UK.
| | | | | | | |
Collapse
|
28
|
Konobu T, Sessler F, Luo LY, Lehmann J. The hNT Human Neuronal Cell Line Survives and Migrates into Rat Retina. Cell Transplant 2017; 7:549-58. [PMID: 9853583 DOI: 10.1177/096368979800700605] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The present studies were undertaken to determine if hNT cells can survive in the vitreous of the eye and migrate into the retina. The hNT neuronal cell line represents a uniform source of human tissue that may be of use in retinal grafts. hNT cells stored in liquid nitrogen were thawed and labeled with the fluorescent dye 1,1′-dioctadecyl-3,3,3′,3′-tetramethylindocarbocyanine Perchlorate (DiI). Thirty thousand cells in 1 μL were injected epiretinally in rat. At survival times of 3, 14, 28, or 56 days, retinal sections were examined quantitatively by epifluorescence to reveal DiI-labeled cells. hNT cells survived in the vitreous at all time points without evidence of vascularization. At 3 days, essentially no hNT cells were found in deep retina, and only very few were attached to retina. At days 14, 28, and 56, hNT cells were found to cluster on the vitreal/retinal interface, and in deeper layers. The clusters of hNT cells took on the shape of a funnel at 14 days, and inverted funnel at 28 days, and by 56 days, populated the photoreceptor layer as a stratum. It is possible that hNT cells took on the morphology and function of photoreceptors. These results suggest that hNT cells injected epiretinally survive in the vitreous at least 56 days, migrate to the retinal/vitreous interface, and may migrate through the retina. This system permits the independent and quantitative evaluation of survival and migratory trophic responses. © 1998 Elsevier Science Inc.
Collapse
Affiliation(s)
- T Konobu
- Department of Neurosurgery, Allegheny University of the Health Sciences, Philadelphia, PA 19102-1192, USA
| | | | | | | |
Collapse
|
29
|
Irons H, Lind JG, Wakade CG, Yu G, Hadman M, Carroll J, Hess DC, Borlongan CV. Intracerebral Xenotransplantation of GFP Mouse Bone Marrow Stromal Cells in Intact and Stroke Rat Brain: Graft Survival and Immunologic Response. Cell Transplant 2017; 13:283-94. [PMID: 15191166 DOI: 10.3727/000000004783983990] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The present study characterized survival and immunologic response of bone marrow stromal cells (BMSCs) following transplantation into intact and stroke brains. In the first study, intrastriatal transplantation of BMSC (60,000 in 3 μl) or vehicle was performed in normal adult Sprague-Dawley male rats that subsequently received daily cyclosporin A (CsA, 10 mg/kg, IP in 3 ml) or vehicle (olive oil, similar volume) starting on day of surgery up to 3 days posttransplantation. Animals were euthanized at 3 or 30 days posttransplantation and brains were processed either for green fluorescent protein (GFP) microscopy or flow cytometry (FACS). Both GFP epifluorescence and FACS scanning revealed GFP+ BMSCs in both groups of transplanted rats with or without CsA, although significantly increased (1.6- to 3-fold more) survival of GFP+ BMSCs was observed in the immunosuppressed animals. Further histologic examination revealed widespread dispersal of BMSCs away from the graft core accompanied by many long outgrowth processes in non-CsA-transplanted animals, whereas a very dense graft core, with cells expressing only sporadic short outgrowth processes, was observed in CsA-transplanted animals. There were no detectable GFP+ BMSCs in nontrans-planted rats that received CsA or vehicle. Immunologic response via FACS analysis revealed a decreased presence of cytotoxic cells, characterized by near complete absence of CD8+ cells, and lack of activation depicted by low CD69 expression in CsA-treated transplanted animals. In contrast, elevated levels of CD8+ cells and increased activation of CD69 expression were observed in transplanted animals that received vehicle alone. CD4+ helper cells were almost nondetectable in transplanted rats that received CsA, but also only minimally elevated in transplanted rats that received vehicle. Nontransplanted rats that received either CsA or vehicle displayed very minimal detectable levels of all three lymphocyte markers. In the second study, a new set of male Sprague-Dawley rats initially received bilateral stereotaxic intrastriatal transplantation of BMSCs and 3 days after were subjected to unilateral transient occlusion of middle cerebral artery. The animals were allowed to survive for 3 days after stroke without CsA immunosuppression. Epifluorescence microscopy revealed significantly higher (5-fold more) survival of transplanted GFP+ BMSCs in the stroke striatum compared with the intact striatum. The majority of the grafts remained within the original dorsal striatal transplant site, characterized by no obvious migration in intact striatum, but with long-distance migration along the ischemic penumbra in the stroke striatum. Moreover, FACS scanning analyses revealed low levels of immunologic response of grafted BMSCs in both stroke and intact striata. These results, taken together, suggest that xenotransplantation of mouse BMSCs into adult rats is feasible. Immunosuppression therapy can enhance xenograft survival and reduce graft-induced immunologic response; however, in the acute phase posttransplantation, BMSCs can survive in intact and stroke brain, and may even exhibit long-distance migration and increased outgrowth processes without immunosuppression.
Collapse
Affiliation(s)
- H Irons
- Department of Neurology, Medical College of Georgia, Augusta, GA 30912, USA
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
Stroke remains a leading cause of death and disability worldwide. An increasing number of animal studies and preclinical trials have, however, provided evidence that regenerative cell-based therapies can lead to functional recovery in stroke patients. Stem cells can differentiate into neural lineages to replace lost neurons. Moreover, they provide trophic support to tissue at risk in the penumbra surrounding the infarct area, enhance vasculogenesis, and help promote survival, migration, and differentiation of the endogenous precursor cells after stroke. Stem cells are highly migratory and seem to be attracted to areas of brain pathology such as ischemic regions. The pathotropism may follow the paradigm of stem cell homing to bone marrow and leukocytes migrating to inflammatory tissue. The molecular signaling therefore may involve various chemokines, cytokines, and integrins. Among these, stromal cell-derived factor-1 (SDF-1)/CXC chemokine receptor-4 (CXCR4) signaling is required for the interaction of stem cells and ischemia-damaged host tissues. SDF-1 is secreted primarily by bone marrow fibroblasts and is required for BMSC homing to bone marrow. Overexpression of SDF-1 in ischemic tissues has been found to enhance stem cell recruitment from peripheral blood and to induce neoangiogenesis. Furthermore, SDF-1 expression in the lesioned area peaked within 7 days postischemia, in concordance with the time window of G-CSF therapy for stroke. Recent data have shown that SDF-1 expression is directly proportional to reduced tissue oxygen tension. SDF-1 gene expression is regulated by hypoxic-inducible factor-1 (HIF-1), a hypoxia-dependent stabilization transcription factor. Thus, ischemic tissue may recruit circulating progenitors regulated by hypoxia through differential expression of HIF-1α and SDF-1. In addition to SDF-1, β2-integrins also play a role in the homing of hematopoietic progenitor cells to sites of ischemia and are critical for their neovascularization capacity. In our recent report, increased expression of β1-integrins apparently contributed to the local neovasculization of the ischemic brain as well as its functional recovery. Identification of the molecular pathways involved in stem cell homing into the ischemic areas could pave the way for the development of new treatment regimens, perhaps using small molecules, designed to enhance endogeneous mobilization of stem cells in various disease states, including chronic stroke and other neurodegenerative diseases. For maximal functional recovery, however, regenerative therapy may need to follow combinatorial approaches, which may include cell replacement, trophic support, protection from oxidative stress, and the neutralization of the growth-inhibitory components for endogenous neuronal stem cells.
Collapse
Affiliation(s)
- Ying-Chao Chang
- Department of Pediatrics, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Chang Guang University College of Medicine, Kaohsiung, Taiwan
| | - Woei-Cherng Shyu
- Neuro-Medical Scientific Center, Tzu-Chi Buddhist General Hospital, Tzu-Chi University, Hualien, Taiwan
| | - Shinn-Zong Lin
- Neuro-Medical Scientific Center, Tzu-Chi Buddhist General Hospital, Tzu-Chi University, Hualien, Taiwan
| | - Hung Li
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
31
|
Fricker-Gates RA, Muir JA, Dunnett SB. Transplanted hNT Cells (“LBS Neurons”) in a Rat Model of Huntington's Disease: Good Survival, Incomplete Differentiation, and Limited Functional Recovery. Cell Transplant 2017; 13:123-36. [PMID: 15129758 DOI: 10.3727/000000004773301807] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
A variety of immortalized cell lines have been proposed to exhibit sufficient phenotypic plasticity to allow them to replace primary embryonic neurons for restorative cell transplantation. In the present experiments we evaluate the functional viability of one particular cell line, the hNT cells developed by Layton Bioscience, to replace lost neurons and alleviate asymmetrical motor deficits in a unilateral excitotoxic lesion model of Huntington's disease. Because the grafts involved implantation of human-derived cells into a rat host environment, all animals were immunosuppressed. Cyclosporin A and FK-506 were similar in providing effective immunoprotection of the hNT xenografts, and whereas the lesions induced a marked inflammatory response in the host brain, this was not exacerbated by the presence of xenograft cells. The presence of grafted cells was determined with the human-specific antigen HuNu, and good graft survival was demonstrated in almost all animals up to the longest survival examined, 16 weeks posttransplantation. Although the cells exhibited progressively greater maturation and differentiation at 10-day, 4- and 16-week time points, staining for the mature neuronal marker NeuN was at best very weak, and we were unable to detect unequivocal staining with any markers of mature striatal phenotype, including DARPP-32, calbindin, parvalbumin, choline acetyl transferase, or NADPH diaphorase (with in all cases positive control provided by good staining on the intact contralateral side of the brain). Nor were we able to detect any differences between rats with lesions alone and rats with grafts in the contralateral motor deficits exhibited in a test of skilled paw reaching or cylinder placing. These results suggest that further and more extensive studies should be undertaken to assess whether hNT neurons can show more extensive and appropriate maturation and be associated with recovery in appropriate behavioral models, before they may be considered a suitable replacement for primary embryonic cells for clinical application in Huntington's disease.
Collapse
|
32
|
Fakoya AOJ. New Delivery Systems of Stem Cells for Vascular Regeneration in Ischemia. Front Cardiovasc Med 2017; 4:7. [PMID: 28286751 PMCID: PMC5323391 DOI: 10.3389/fcvm.2017.00007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Accepted: 02/07/2017] [Indexed: 01/08/2023] Open
Abstract
The finances of patients and countries are increasingly overwhelmed with the plague of cardiovascular diseases as a result of having to chronically manage the associated complications of ischemia such as heart failures, neurological deficits, chronic limb ulcers, gangrenes, and amputations. Hence, scientific research has sought for alternate therapies since pharmacological and surgical treatments have fallen below expectations in providing the desired quality of life. The advent of stem cells research has raised expectations with respect to vascular regeneration and tissue remodeling, hence assuring the patients of the possibility of an improved quality of life. However, these supposed encouraging results have been short-lived as the retention, survival, and engraftment rates of these cells appear to be inadequate; hence, the long-term beneficial effects of these cells cannot be ascertained. These drawbacks have led to the relentless research into better ways to deliver stem cells or angiogenic factors (which mobilize stem cells) to the regions of interest to facilitate increased retention, survival, engraftment, and regeneration. This review considered methods, such as the use of scaffolds, retrograde coronary delivery, improved combinations, stem cell pretreatment, preconditioning, stem cell exosomes, mannitol, magnet, and ultrasound-enhanced delivery, homing techniques, and stem cell modulation. Furthermore, the study appraised the possibility of a combination therapy of stem cells and macrophages, considering the enormous role macrophages play in repair, remodeling, and angiogenesis.
Collapse
|
33
|
Limb Remote Ischemic Postconditioning Reduces Ischemia-Reperfusion Injury by Inhibiting NADPH Oxidase Activation and MyD88-TRAF6-P38MAP-Kinase Pathway of Neutrophils. Int J Mol Sci 2016; 17:ijms17121971. [PMID: 27898007 PMCID: PMC5187771 DOI: 10.3390/ijms17121971] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 10/30/2016] [Accepted: 11/17/2016] [Indexed: 12/11/2022] Open
Abstract
Limb remote ischemic postconditioning (LRIP) has been confirmed to reduce the ischemia-reperfusion injury but its mechanisms are still not clear. This study clarified the mechanism of LRIP based on the nicotinamide-adenine dinucleotide phosphate (NADPH) oxidase and Myeloid differentiation factor 88 (MyD88)-Tumor necrosis factor (TNF) receptor-associated factor 6 (TRAF6)-P38 pathway of neutrophils. Rat middle cerebral artery occlusion (MCAO) model was used in this study. Ischemia-reperfusion injury was carried out by MCAO 1.5 h followed by 24 h reperfusion. LRIP operation was performed to the left femoral artery at 0, 1 or 3 h after reperfusion. Behavioral testing, including postural reflex test, vibrissae-elicited forelimb placing test and tail hang test, showed that LRIP operated at 0 h of reperfusion could significantly ameliorate these behavioral scores. Pathological examinations, infarct size, Myeloperoxidase (MPO) activity showed that LRIP operated at 0 h of reperfusion could significantly ameliorate the pathological scores, reduce the infarct size and MPO activity in the brain and increase the MPO activity in the left leg. By using Neutrophil counting, immunofluorescence and real-time PCR techniques, we found that LRIP operated at 0 h of reperfusion could reduce neutrophil counts in the peripheral blood and downregulate the activation of neutrophil in the peripheral blood and rat brain. Western blots revealed that MyD88, TRAF6, p38 mitogen-activated protein kinase (p38-MAPK) in neutrophils and the phosphorylation of p47phox (Ser 304 and Ser 345) in neutrophil could be downregulated by LRIP. Our study suggests that LRIP inhibits the number and activation of neutrophils in the rat brain and peripheral blood linked to down-regulating the activation of NADPH oxidase in neutrophils by MyD88/TRAF6/p38-MAPK pathway.
Collapse
|
34
|
Uhlendorf TL, Nuryyev RL, Kopyov AO, Ochoa J, Younesi S, Cohen RW, Kopyov OV. Efficacy of Two Delivery Routes for Transplanting Human Neural Progenitor Cells (NPCs) Into the Spastic Han-Wistar Rat, a Model of Ataxia. Cell Transplant 2016; 26:259-269. [PMID: 27938495 DOI: 10.3727/096368916x693527] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
An emerging avenue for recalcitrant neurodegenerative disease treatment is neural progenitor cell (NPC) transplantation. In this study, we investigated the effectiveness of two different delivery routes of human-derived NPC inoculation: injection into the common carotid artery or unilateral stereotactic implantation into the degenerating cerebellum and hippocampus of spastic Han-Wistar (sHW) rats, a model of ataxia. At 30 days of age, sHW mutants were implanted with osmotic pumps preloaded with cyclosporine. Ten days after pump implantation, the animals were given either 3,000,000 live human-derived NPCs (hNPCs; n = 12) or 3,000,000 dead NPCs (dNPCs; n = 12) injected into the common carotid artery, or were given two unilateral implantations of 500,000 hNPCs into the cerebellum and 500,000 hNPCs into the hippocampus of each sHW rat (n = 12) or 500,000 dNPCs by unilateral implantation into the cerebellum and hippocampus (n = 12). We also compared treated sHW rats to untreated sHW rats: normal rats (n = 12) and sibling sHW rats (n = 12). Motor activity and animal weights were monitored every 5 days to ascertain effectiveness of the two types of delivery methods compared to the untreated mutant and normal animals. Mutant rats with hNPC implantations, but not dNPC or carotid artery injections, showed significant deceleration of motor deterioration (p < 0.05). These mutants with hNPC implantations also retained weight longer than dNPC mutants did (p < 0.05). At the end of the experiment, animals were sacrificed for histological evaluation. Using fluorescent markers (Qtracker) incorporated into the hNPC prior to implantation and human nuclear immunostaining, we observed few hNPCs in the brains of carotid artery-injected mutants. However, significant numbers of surviving hNPCs were seen using these techniques in mutant cerebellums and hippocampi implanted with hNPC. Our results show that direct implantation of hNPCs reduced ataxic symptoms in the sHW rat, demonstrating that stereotactic route of stem cell delivery correlates to improved clinical outcomes.
Collapse
|
35
|
Wang JY, Huang YN, Chiu CC, Tweedie D, Luo W, Pick CG, Chou SY, Luo Y, Hoffer BJ, Greig NH, Wang JY. Pomalidomide mitigates neuronal loss, neuroinflammation, and behavioral impairments induced by traumatic brain injury in rat. J Neuroinflammation 2016; 13:168. [PMID: 27353053 PMCID: PMC4924242 DOI: 10.1186/s12974-016-0631-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 06/16/2016] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) is a global health concern that typically causes emotional disturbances and cognitive dysfunction. Secondary pathologies following TBI may be associated with chronic neurodegenerative disorders and an enhanced likelihood of developing dementia-like disease in later life. There are currently no approved drugs for mitigating the acute or chronic effects of TBI. METHODS The effects of the drug pomalidomide (Pom), an FDA-approved immunomodulatory agent, were evaluated in a rat model of moderate to severe TBI induced by controlled cortical impact. Post-TBI intravenous administration of Pom (0.5 mg/kg at 5 or 7 h and 0.1 mg/kg at 5 h) was evaluated on functional and histological measures that included motor function, fine more coordination, somatosensory function, lesion volume, cortical neurodegeneration, neuronal apoptosis, and the induction of pro-inflammatory cytokines (TNF-α, IL-1β, IL-6). RESULTS Pom 0.5 mg/kg administration at 5 h, but not at 7 h post-TBI, significantly mitigated the TBI-induced injury volume and functional impairments, neurodegeneration, neuronal apoptosis, and cytokine mRNA and protein induction. To evaluate underlying mechanisms, the actions of Pom on neuronal survival, microglial activation, and the induction of TNF-α were assessed in mixed cortical cultures following a glutamate challenge. Pom dose-dependently ameliorated glutamate-mediated cytotoxic effects on cell viability and reduced microglial cell activation, significantly attenuating the induction of TNF-α. CONCLUSIONS Post-injury treatment with a single Pom dose within 5 h significantly reduced functional impairments in a well-characterized animal model of TBI. Pom decreased the injury lesion volume, augmented neuronal survival, and provided anti-inflammatory properties. These findings strongly support the further evaluation and optimization of Pom for potential use in clinical TBI.
Collapse
Affiliation(s)
- Jin-Ya Wang
- Graduate Institute of Medical Science, College of Medicine, Taipei Medical University, 250 Wu-Hsing St., Taipei, 110 Taiwan
| | - Ya-Ni Huang
- Graduate Institute of Medical Science, College of Medicine, Taipei Medical University, 250 Wu-Hsing St., Taipei, 110 Taiwan
- Department of Nursing, Hsin Sheng Junior College of Medical Care and Management, Taoyuan, Taiwan
| | - Chong-Chi Chiu
- Department of General Surgery, Chi Mei Medical Center, Tainan and Liouying, Taiwan
| | - David Tweedie
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, USA
| | - Weiming Luo
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, USA
| | - Chaim G. Pick
- Department of Anatomy and Anthropology, Sackler School of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Szu-Yi Chou
- Graduate Program on Neuroregeneration, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Yu Luo
- Department of Neurosurgery, Case Western Reserve University School of Medicine, Cleveland, OH USA
| | - Barry J. Hoffer
- Department of Neurosurgery, Case Western Reserve University School of Medicine, Cleveland, OH USA
| | - Nigel H. Greig
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, USA
| | - Jia-Yi Wang
- Graduate Institute of Medical Science, College of Medicine, Taipei Medical University, 250 Wu-Hsing St., Taipei, 110 Taiwan
- Department of Physiology, College of Medicine, Taipei Medical University, 250 Wu-Hsing St., Taipei, 110 Taiwan
| |
Collapse
|
36
|
Ajioka I. Biomaterial-engineering and neurobiological approaches for regenerating the injured cerebral cortex. Regen Ther 2016; 3:63-67. [PMID: 31245474 PMCID: PMC6581816 DOI: 10.1016/j.reth.2016.02.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 01/08/2016] [Accepted: 02/12/2016] [Indexed: 01/07/2023] Open
Abstract
The cerebral cortex is responsible for higher functions of the central nervous system (CNS), such as movement, sensation, and cognition. When the cerebral cortex is severely injured, these functions are irreversibly impaired. Although recent neurobiological studies reveal that the cortex has the potential for regeneration, therapies for functional recovery face some technological obstacles. Biomaterials have been used to evoke regenerative potential and promote regeneration in several tissues, including the CNS. This review presents a brief overview of new therapeutic strategies for cortical regeneration from the perspectives of neurobiology and biomaterial engineering, and discusses a promising technology for evoking the regenerative potential of the cerebral cortex.
Collapse
Affiliation(s)
- Itsuki Ajioka
- Center for Brain Integration Research (CBIR), Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan,The Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan,Center for Brain Integration Research, Tokyo Medical and Dental University, 1-5-45 Yushima, Tokyo 113-8510, Japan. Fax: +81 3 5803 4716.
| |
Collapse
|
37
|
González-Burguera I, Ricobaraza A, Aretxabala X, Barrondo S, García del Caño G, López de Jesús M, Sallés J. Highly efficient generation of glutamatergic/cholinergic NT2-derived postmitotic human neurons by short-term treatment with the nucleoside analogue cytosine β-D-arabinofuranoside. Stem Cell Res 2016; 16:541-51. [PMID: 26985738 DOI: 10.1016/j.scr.2016.02.038] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 01/25/2016] [Accepted: 02/26/2016] [Indexed: 12/21/2022] Open
Abstract
The human NTERA2/D1 (NT2) cells generate postmitotic neurons (NT2N cells) upon retinoic acid (RA) treatment and are functionally integrated in the host tissue following grafting into the rodent and human brain, thus representing a promising source for neuronal replacement therapy. Yet the major limitations of this model are the lengthy differentiation procedure and its low efficiency, although recent studies suggest that the differentiation process can be shortened to less than 1 week using nucleoside analogues. To explore whether short-term exposure of NT2 cells to the nucleoside analogue cytosine β-d-arabinofuranoside (AraC) could be a suitable method to efficiently generate mature neurons, we conducted a neurochemical and morphometric characterization of AraC-differentiated NT2N (AraC/NT2N) neurons and improved the differentiation efficiency by modifying the cell culture schedule. Moreover, we analyzed the neurotransmitter phenotypes of AraC/NT2N neurons. Cultures obtained by treatment with AraC were highly enriched in postmitotic neurons and essentially composed of dual glutamatergic/cholinergic neurons, which contrasts with the preferential GABAergic phenotype that we found after RA differentiation. Taken together, our results further reinforce the notion NT2 cells are a versatile source of neuronal phenotypes and provide a new encouraging platform for studying mechanisms of neuronal differentiation and for exploring neuronal replacement strategies.
Collapse
Affiliation(s)
- Imanol González-Burguera
- Department of Pharmacology, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz (Araba), Spain.
| | - Ana Ricobaraza
- Department of Pharmacology, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz (Araba), Spain.
| | - Xabier Aretxabala
- Department of Neurosciences, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz (Araba), Spain.
| | - Sergio Barrondo
- Department of Pharmacology, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz (Araba), Spain; CIBERSAM, Spain.
| | - Gontzal García del Caño
- Department of Neurosciences, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz (Araba), Spain.
| | - Maider López de Jesús
- Department of Pharmacology, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz (Araba), Spain; CIBERSAM, Spain.
| | - Joan Sallés
- Department of Pharmacology, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz (Araba), Spain; CIBERSAM, Spain.
| |
Collapse
|
38
|
Ingberg E, Gudjonsdottir J, Theodorsson E, Theodorsson A, Ström JO. Elevated body swing test after focal cerebral ischemia in rodents: methodological considerations. BMC Neurosci 2015; 16:50. [PMID: 26242584 PMCID: PMC4525734 DOI: 10.1186/s12868-015-0189-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 07/28/2015] [Indexed: 01/06/2023] Open
Abstract
Background The elevated body swing test (EBST) is a behavioral test used to evaluate experimental stroke in rodents. The basic idea is that when the animal is suspended vertically by the tail, it will swing its head laterally to the left or right depending on lesion side. In a previous study from our lab using the EBST after middle cerebral artery occlusion (MCAo), rats swung contralateral to the infarct day 1 post-MCAo, but ipsilateral day 3 post-MCAo. This shift was unexpected and prompted us to perform the present study. First, the literature was systematically reviewed to elucidate whether a similar shift had been noticed before, and if consensus existed regarding swing direction. Secondly, an experiment was conducted to systematically investigate the suggested behavior. Eighty-three adult male and female Sprague–Dawley rats were subjected to MCAo or sham surgery and the EBST was performed up to 7 days after the lesion. Results Both experimentally and through systematic literature review, the present study shows that the direction of biased swing activity in the EBST for rodents after cerebral ischemia can differ and even shift over time in some situations. The EBST curve for females was significantly different from that of males after the same occlusion time (p = 0.023). Conclusions This study highlights the importance of adequate reporting of behavioral tests for lateralization and it is concluded that the EBST cannot be recommended as a test for motor asymmetry after MCAo in rats.
Collapse
Affiliation(s)
- Edvin Ingberg
- Division of Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Department of Clinical Chemistry, Faculty of Health Sciences, Center for Diagnostics, Linköping University, Region Östergötland, Linköping, Sweden.
| | - Johanna Gudjonsdottir
- Division of Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Department of Clinical Chemistry, Faculty of Health Sciences, Center for Diagnostics, Linköping University, Region Östergötland, Linköping, Sweden.
| | - Elvar Theodorsson
- Division of Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Department of Clinical Chemistry, Faculty of Health Sciences, Center for Diagnostics, Linköping University, Region Östergötland, Linköping, Sweden.
| | - Annette Theodorsson
- Division of Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Department of Clinical Chemistry, Faculty of Health Sciences, Center for Diagnostics, Linköping University, Region Östergötland, Linköping, Sweden. .,Division of Neuroscience, Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Department of Neurosurgery, Anaesthetics, Operations and Specialty Surgery Center, Linköping University, Region Östergötland, Linköping, Sweden.
| | - Jakob O Ström
- Division of Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Department of Clinical Chemistry, Faculty of Health Sciences, Center for Diagnostics, Linköping University, Region Östergötland, Linköping, Sweden. .,Vårdvetenskapligt Forskningscentrum/Centre for Health Sciences, Örebro University Hospital, County Council of Örebro, Örebro, Sweden. .,School of Health and Medical Sciences, Örebro University, Örebro, Sweden.
| |
Collapse
|
39
|
Janowski M, Wagner DC, Boltze J. Stem Cell-Based Tissue Replacement After Stroke: Factual Necessity or Notorious Fiction? Stroke 2015; 46:2354-63. [PMID: 26106118 DOI: 10.1161/strokeaha.114.007803] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 04/28/2015] [Indexed: 01/09/2023]
Affiliation(s)
- Miroslaw Janowski
- From the Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research (M.J.) and Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering (M.J.), The Johns Hopkins University School of Medicine, Baltimore, MD; NeuroRepair Department (M.J.) and Department of Neurosurgery (M.J.), Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland; Department of Cell Therapy, Fraunhofer-Institute for Cell Therapy and Immunology, Translational Centre for Regenerative Medicine, Leipzig, Germany (D.-C.W., J.B.); and Stroke and Neurovascular Regulation Laboratory, Neuroscience Center at Massachussets General Hospital, Harvard Medical School, Stroke and Neurovascular Regulation Laboratory, Charlestown, MA (J.B.)
| | - Daniel-Christoph Wagner
- From the Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research (M.J.) and Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering (M.J.), The Johns Hopkins University School of Medicine, Baltimore, MD; NeuroRepair Department (M.J.) and Department of Neurosurgery (M.J.), Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland; Department of Cell Therapy, Fraunhofer-Institute for Cell Therapy and Immunology, Translational Centre for Regenerative Medicine, Leipzig, Germany (D.-C.W., J.B.); and Stroke and Neurovascular Regulation Laboratory, Neuroscience Center at Massachussets General Hospital, Harvard Medical School, Stroke and Neurovascular Regulation Laboratory, Charlestown, MA (J.B.)
| | - Johannes Boltze
- From the Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research (M.J.) and Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering (M.J.), The Johns Hopkins University School of Medicine, Baltimore, MD; NeuroRepair Department (M.J.) and Department of Neurosurgery (M.J.), Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland; Department of Cell Therapy, Fraunhofer-Institute for Cell Therapy and Immunology, Translational Centre for Regenerative Medicine, Leipzig, Germany (D.-C.W., J.B.); and Stroke and Neurovascular Regulation Laboratory, Neuroscience Center at Massachussets General Hospital, Harvard Medical School, Stroke and Neurovascular Regulation Laboratory, Charlestown, MA (J.B.).
| |
Collapse
|
40
|
Affiliation(s)
- Theo Diamandis
- From the Department of Neurosugery and Brain Repair, Center of Excellence for Aging and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa
| | - Cesar V Borlongan
- From the Department of Neurosugery and Brain Repair, Center of Excellence for Aging and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa.
| |
Collapse
|
41
|
Christie SD, Sadi D, Mendez I. Intraspinal Transplantation of hNT Neurons in the Lesioned Adult Rat Spinal Cord. Can J Neurol Sci 2014; 31:87-96. [PMID: 15038476 DOI: 10.1017/s0317167100002882] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Background:The role of neural transplantation as a restorative strategy for spinal cord injury continues to be intensely investigated. Ideally, the tissue source for transplantation must be readily available, free of disease and able to survive and mature following implantation into the adverse environment created by the injury. We have studied the use of a commercially available cell line of cultured human neurons (hNT neurons) as a tissue source for neural transplantation in spinal cord injury.Methods:Following a left lateral thoracic hemisection, 54 immunosuppressed, female Wistar rats were randomly allocated into different treatment groups; hemisection only or hemisection and hNT cell transplantation (via a bridge, double or triple graft). Grafting occurred three days after spinal cord injury. After thirteen weeks the animals were sacrificed and tissue sections were stained with human neuron specific enolase and human specific neural cell adhesion molecule.Results:Immunohistochemical evidence of graft survival was displayed in 66.7% of the surviving, grafted animals. Fibre outgrowth, greatest in the bridge and triple grafts, was observed in both rostral and caudal directions essentially bridging the lesion. Double grafts were smaller, displaying less fibre outgrowth, which did not cross the lesion. Long fibre outgrowth was evident up to 2 cm from the graft as assessed by tracing and immunohistochemical studies.Conclusion:Bridge and triple grafts displayed greater growth and enabled the hNT graft to essentially bridge the lesion. This suggests that hNT neurons have the potential to structurally reconnect the proximal and distal spinal cord across the region of injury.
Collapse
Affiliation(s)
- Sean Dennis Christie
- Department of Surgery (Neurosurgery), Dalhousie University, Halifax, Nova Scotia, Canada
| | | | | |
Collapse
|
42
|
Detante O, Jaillard A, Moisan A, Barbieux M, Favre I, Garambois K, Hommel M, Remy C. Biotherapies in stroke. Rev Neurol (Paris) 2014; 170:779-98. [DOI: 10.1016/j.neurol.2014.10.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 09/29/2014] [Accepted: 10/08/2014] [Indexed: 12/31/2022]
|
43
|
Gonzales-Portillo GS, Reyes S, Aguirre D, Pabon MM, Borlongan CV. Stem cell therapy for neonatal hypoxic-ischemic encephalopathy. Front Neurol 2014; 5:147. [PMID: 25161645 PMCID: PMC4130306 DOI: 10.3389/fneur.2014.00147] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2014] [Accepted: 07/22/2014] [Indexed: 11/27/2022] Open
Abstract
Treatments for neonatal hypoxic-ischemic encephalopathy (HIE) have been limited. The aim of this paper is to offer translational research guidance on stem cell therapy for neonatal HIE by examining clinically relevant animal models, practical stem cell sources, safety and efficacy of endpoint assays, as well as a general understanding of modes of action of this cellular therapy. In order to do so, we discuss the clinical manifestations of HIE, highlighting its overlapping pathologies with stroke and providing insights on the potential of cell therapy currently investigated in stroke, for HIE. To this end, we draw guidance from recommendations outlined in stem cell therapeutics as an emerging paradigm for stroke or STEPS, which have been recently modified to Baby STEPS to cater for the “neonatal” symptoms of HIE. These guidelines recognized that neonatal HIE exhibit distinct disease symptoms from adult stroke in need of an innovative translational approach that facilitates the entry of cell therapy in the clinic. Finally, new information about recent clinical trials and insights into combination therapy are provided with the vision that stem cell therapy may benefit from available treatments, such as hypothermia, already being tested in children diagnosed with HIE.
Collapse
Affiliation(s)
| | - Stephanny Reyes
- Department of Neurosurgery and Brain Repair, University of South Florida , Tampa, FL , USA
| | - Daniela Aguirre
- Department of Neurosurgery and Brain Repair, University of South Florida , Tampa, FL , USA
| | - Mibel M Pabon
- Department of Neurosurgery and Brain Repair, University of South Florida , Tampa, FL , USA
| | - Cesar V Borlongan
- Department of Neurosurgery and Brain Repair, University of South Florida , Tampa, FL , USA
| |
Collapse
|
44
|
Banerjee S, Bentley P, Hamady M, Marley S, Davis J, Shlebak A, Nicholls J, Williamson DA, Jensen SL, Gordon M, Habib N, Chataway J. Intra-Arterial Immunoselected CD34+ Stem Cells for Acute Ischemic Stroke. Stem Cells Transl Med 2014; 3:1322-30. [PMID: 25107583 DOI: 10.5966/sctm.2013-0178] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Treatment with CD34+ hematopoietic stem/progenitor cells has been shown to improve functional recovery in nonhuman models of ischemic stroke via promotion of angiogenesis and neurogenesis. We aimed to determine the safety and feasibility of treatment with CD34+ cells delivered intra-arterially in patients with acute ischemic stroke. This was the first study in human subjects. We performed a prospective, nonrandomized, open-label, phase I study of autologous, immunoselected CD34+ stem/progenitor cell therapy in patients presenting within 7 days of onset with severe anterior circulation ischemic stroke (National Institutes of Health Stroke Scale [NIHSS] score≥8). CD34+ cells were collected from the bone marrow of the subjects before being delivered by catheter angiography into the ipsilesional middle cerebral artery. Eighty-two patients with severe anterior circulation ischemic stroke were screened, of whom five proceeded to treatment. The common reasons for exclusion were age>80 years (n=19); medical instability (n=17), and significant carotid stenosis (n=13). The procedure was well tolerated in all patients, and no significant treatment-related adverse effects occurred. All patients showed improvements in clinical functional scores (Modified Rankin Score and NIHSS score) and reductions in lesion volume during a 6-month follow-up period. Autologous CD34+ selected stem/progenitor cell therapy delivered intra-arterially into the infarct territory can be achieved safely in patients with acute ischemic stroke. Future studies that address eligibility criteria, dosage, delivery site, and timing and that use surrogate imaging markers of outcome are desirable before larger scale clinical trials.
Collapse
Affiliation(s)
- Soma Banerjee
- Department of Stroke Medicine, Clinical Neurosciences, Department of Interventional Radiology, Stem Cell Transplant Unit, and Department of Haematology, Imperial College Healthcare National Health Services Trust, London, United Kingdom; Departments of Surgery and Haematology, Faculty of Medicine, Imperial College London, London, United Kingdom; Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand; National Hospital for Neurology and Neurosurgery, University College London Hospitals National Health Services Foundation Trust, London, United Kingdom; Institute of Neurology, University College London, London, United Kingdom
| | - Paul Bentley
- Department of Stroke Medicine, Clinical Neurosciences, Department of Interventional Radiology, Stem Cell Transplant Unit, and Department of Haematology, Imperial College Healthcare National Health Services Trust, London, United Kingdom; Departments of Surgery and Haematology, Faculty of Medicine, Imperial College London, London, United Kingdom; Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand; National Hospital for Neurology and Neurosurgery, University College London Hospitals National Health Services Foundation Trust, London, United Kingdom; Institute of Neurology, University College London, London, United Kingdom
| | - Mohammad Hamady
- Department of Stroke Medicine, Clinical Neurosciences, Department of Interventional Radiology, Stem Cell Transplant Unit, and Department of Haematology, Imperial College Healthcare National Health Services Trust, London, United Kingdom; Departments of Surgery and Haematology, Faculty of Medicine, Imperial College London, London, United Kingdom; Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand; National Hospital for Neurology and Neurosurgery, University College London Hospitals National Health Services Foundation Trust, London, United Kingdom; Institute of Neurology, University College London, London, United Kingdom
| | - Stephen Marley
- Department of Stroke Medicine, Clinical Neurosciences, Department of Interventional Radiology, Stem Cell Transplant Unit, and Department of Haematology, Imperial College Healthcare National Health Services Trust, London, United Kingdom; Departments of Surgery and Haematology, Faculty of Medicine, Imperial College London, London, United Kingdom; Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand; National Hospital for Neurology and Neurosurgery, University College London Hospitals National Health Services Foundation Trust, London, United Kingdom; Institute of Neurology, University College London, London, United Kingdom
| | - John Davis
- Department of Stroke Medicine, Clinical Neurosciences, Department of Interventional Radiology, Stem Cell Transplant Unit, and Department of Haematology, Imperial College Healthcare National Health Services Trust, London, United Kingdom; Departments of Surgery and Haematology, Faculty of Medicine, Imperial College London, London, United Kingdom; Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand; National Hospital for Neurology and Neurosurgery, University College London Hospitals National Health Services Foundation Trust, London, United Kingdom; Institute of Neurology, University College London, London, United Kingdom
| | - Abdul Shlebak
- Department of Stroke Medicine, Clinical Neurosciences, Department of Interventional Radiology, Stem Cell Transplant Unit, and Department of Haematology, Imperial College Healthcare National Health Services Trust, London, United Kingdom; Departments of Surgery and Haematology, Faculty of Medicine, Imperial College London, London, United Kingdom; Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand; National Hospital for Neurology and Neurosurgery, University College London Hospitals National Health Services Foundation Trust, London, United Kingdom; Institute of Neurology, University College London, London, United Kingdom
| | - Joanna Nicholls
- Department of Stroke Medicine, Clinical Neurosciences, Department of Interventional Radiology, Stem Cell Transplant Unit, and Department of Haematology, Imperial College Healthcare National Health Services Trust, London, United Kingdom; Departments of Surgery and Haematology, Faculty of Medicine, Imperial College London, London, United Kingdom; Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand; National Hospital for Neurology and Neurosurgery, University College London Hospitals National Health Services Foundation Trust, London, United Kingdom; Institute of Neurology, University College London, London, United Kingdom
| | - Deborah A Williamson
- Department of Stroke Medicine, Clinical Neurosciences, Department of Interventional Radiology, Stem Cell Transplant Unit, and Department of Haematology, Imperial College Healthcare National Health Services Trust, London, United Kingdom; Departments of Surgery and Haematology, Faculty of Medicine, Imperial College London, London, United Kingdom; Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand; National Hospital for Neurology and Neurosurgery, University College London Hospitals National Health Services Foundation Trust, London, United Kingdom; Institute of Neurology, University College London, London, United Kingdom
| | - Steen L Jensen
- Department of Stroke Medicine, Clinical Neurosciences, Department of Interventional Radiology, Stem Cell Transplant Unit, and Department of Haematology, Imperial College Healthcare National Health Services Trust, London, United Kingdom; Departments of Surgery and Haematology, Faculty of Medicine, Imperial College London, London, United Kingdom; Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand; National Hospital for Neurology and Neurosurgery, University College London Hospitals National Health Services Foundation Trust, London, United Kingdom; Institute of Neurology, University College London, London, United Kingdom
| | - Myrtle Gordon
- Department of Stroke Medicine, Clinical Neurosciences, Department of Interventional Radiology, Stem Cell Transplant Unit, and Department of Haematology, Imperial College Healthcare National Health Services Trust, London, United Kingdom; Departments of Surgery and Haematology, Faculty of Medicine, Imperial College London, London, United Kingdom; Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand; National Hospital for Neurology and Neurosurgery, University College London Hospitals National Health Services Foundation Trust, London, United Kingdom; Institute of Neurology, University College London, London, United Kingdom
| | - Nagy Habib
- Department of Stroke Medicine, Clinical Neurosciences, Department of Interventional Radiology, Stem Cell Transplant Unit, and Department of Haematology, Imperial College Healthcare National Health Services Trust, London, United Kingdom; Departments of Surgery and Haematology, Faculty of Medicine, Imperial College London, London, United Kingdom; Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand; National Hospital for Neurology and Neurosurgery, University College London Hospitals National Health Services Foundation Trust, London, United Kingdom; Institute of Neurology, University College London, London, United Kingdom
| | - Jeremy Chataway
- Department of Stroke Medicine, Clinical Neurosciences, Department of Interventional Radiology, Stem Cell Transplant Unit, and Department of Haematology, Imperial College Healthcare National Health Services Trust, London, United Kingdom; Departments of Surgery and Haematology, Faculty of Medicine, Imperial College London, London, United Kingdom; Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand; National Hospital for Neurology and Neurosurgery, University College London Hospitals National Health Services Foundation Trust, London, United Kingdom; Institute of Neurology, University College London, London, United Kingdom
| |
Collapse
|
45
|
Abstract
On average, every four minutes an individual dies from a stroke, accounting for 1 out of every 18 deaths in the United States. Approximately 795,000 Americans have a new or recurrent stroke each year, with just over 600,000 of these being first attack [1]. There have been multiple animal models of stroke demonstrating that novel therapeutics can help improve the clinical outcome. However, these results have failed to show the same outcomes when tested in human clinical trials. This review will discuss the current in vivo animal models of stroke, advantages and limitations, and the rationale for employing these animal models to satisfy translational gating items for examination of neuroprotective, as well as neurorestorative strategies in stroke patients. An emphasis in the present discussion of therapeutics development is given to stem cell therapy for stroke.
Collapse
|
46
|
Stem cell therapy for acute cerebral injury: what do we know and what will the future bring? Curr Opin Neurol 2014; 26:617-25. [PMID: 24136128 DOI: 10.1097/wco.0000000000000023] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
PURPOSE OF REVIEW The central nervous system has limited capacity for regeneration after acute and chronic injury. An attractive approach to stimulate neural plasticity in the brain is to transplant stem cells in order to restore function. Here, we discuss potential mechanisms of action, current knowledge and future perspectives of clinical stem cell research for stroke and traumatic brain injury. RECENT FINDINGS Preclinical data using various models suggest stem cell therapy to be a promising therapeutic avenue. Progress has been made in elucidating the mechanism of action of various cell types used, shifting the hypothesis from neural replacement to enhancing endogenous repair processes. Translation of these findings in clinical trials is currently being pursued with emphasis on both safety as well as efficacy. SUMMARY Clinical trials are currently recruiting patients in phase I and II trials to gain more insight in the therapeutic potential of stem cells in acute cerebral injury. A close interplay between results of these clinical trials and more extensive basic research is essential for future trial design, choosing the optimal transplantation strategy and selecting the right patients.
Collapse
|
47
|
Vishwakarma SK, Bardia A, Tiwari SK, Paspala SA, Khan AA. Current concept in neural regeneration research: NSCs isolation, characterization and transplantation in various neurodegenerative diseases and stroke: A review. J Adv Res 2014; 5:277-94. [PMID: 25685495 PMCID: PMC4294738 DOI: 10.1016/j.jare.2013.04.005] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 04/10/2013] [Accepted: 04/28/2013] [Indexed: 12/14/2022] Open
Abstract
Since last few years, an impressive amount of data has been generated regarding the basic in vitro and in vivo biology of neural stem cells (NSCs) and there is much far hope for the success in cell replacement therapies for several human neurodegenerative diseases and stroke. The discovery of adult neurogenesis (the endogenous production of new neurons) in the mammalian brain more than 40 years ago has resulted in a wealth of knowledge about stem cells biology in neuroscience research. Various studies have done in search of a suitable source for NSCs which could be used in animal models to understand the basic and transplantation biology before treating to human. The difficulties in isolating pure population of NSCs limit the study of neural stem behavior and factors that regulate them. Several studies on human fetal brain and spinal cord derived NSCs in animal models have shown some interesting results for cell replacement therapies in many neurodegenerative diseases and stroke models. Also the methods and conditions used for in vitro culture of these cells provide an important base for their applicability and specificity in a definite target of the disease. Various important developments and modifications have been made in stem cells research which is needed to be more specified and enrolment in clinical studies using advanced approaches. This review explains about the current perspectives and suitable sources for NSCs isolation, characterization, in vitro proliferation and their use in cell replacement therapies for the treatment of various neurodegenerative diseases and strokes.
Collapse
Affiliation(s)
- Sandeep K. Vishwakarma
- Centre for Liver Research and Diagnostics, Deccan College of Medical Sciences, Kanchanbagh, Hyderabad, 500 058 Andhra Pradesh, India
- Paspala Advanced Neural (PAN) Research Foundation, Narayanguda, Hyderabad, 500 029 Andhra Pradesh, India
| | - Avinash Bardia
- Centre for Liver Research and Diagnostics, Deccan College of Medical Sciences, Kanchanbagh, Hyderabad, 500 058 Andhra Pradesh, India
| | - Santosh K. Tiwari
- Centre for Liver Research and Diagnostics, Deccan College of Medical Sciences, Kanchanbagh, Hyderabad, 500 058 Andhra Pradesh, India
| | - Syed A.B. Paspala
- Centre for Liver Research and Diagnostics, Deccan College of Medical Sciences, Kanchanbagh, Hyderabad, 500 058 Andhra Pradesh, India
- Paspala Advanced Neural (PAN) Research Foundation, Narayanguda, Hyderabad, 500 029 Andhra Pradesh, India
| | - Aleem A. Khan
- Centre for Liver Research and Diagnostics, Deccan College of Medical Sciences, Kanchanbagh, Hyderabad, 500 058 Andhra Pradesh, India
- Paspala Advanced Neural (PAN) Research Foundation, Narayanguda, Hyderabad, 500 029 Andhra Pradesh, India
| |
Collapse
|
48
|
Ectopic expression of human angiopoietin-1 promotes functional recovery and neurogenesis after focal cerebral ischemia. Neuroscience 2014; 267:135-46. [DOI: 10.1016/j.neuroscience.2014.02.036] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 01/28/2014] [Accepted: 02/24/2014] [Indexed: 11/22/2022]
|
49
|
Tajiri N, Quach DM, Kaneko Y, Wu S, Lee D, Lam T, Hayama KL, Hazel TG, Johe K, Wu MC, Borlongan CV. Behavioral and histopathological assessment of adult ischemic rat brains after intracerebral transplantation of NSI-566RSC cell lines. PLoS One 2014; 9:e91408. [PMID: 24614895 PMCID: PMC3948841 DOI: 10.1371/journal.pone.0091408] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 02/10/2014] [Indexed: 02/06/2023] Open
Abstract
Stroke is a major cause of death and disability, with very limited treatment option. Cell-based therapies have emerged as potential treatments for stroke. Indeed, studies have shown that transplantation of neural stem cells (NSCs) exerts functional benefits in stroke models. However, graft survival and integration with the host remain pressing concerns with cell-based treatments. The current study set out to investigate those very issues using a human NSC line, NSI-566RSC, in a rat model of ischemic stroke induced by transient occlusion of the middle cerebral artery. Seven days after stroke surgery, those animals that showed significant motor and neurological impairments were randomly assigned to receive NSI-566RSC intracerebral transplants at two sites within the striatum at three different doses: group A (0 cells/µl), group B (5,000 cells/µl), group C (10,000 cells/µl), and group D (20,000 cells/µl). Weekly behavioral tests, starting at seven days and continued up to 8 weeks after transplantation, revealed dose-dependent recovery from both motor and neurological deficits in transplanted stroke animals. Eight weeks after cell transplantation, immunohistochemical investigations via hematoxylin and eosin staining revealed infarct size was similar across all groups. To identify the cell graft, and estimate volume, immunohistochemistry was performed using two human-specific antibodies: one to detect all human nuclei (HuNu), and another to detect human neuron-specific enolase (hNSE). Surviving cell grafts were confirmed in 10/10 animals of group B, 9/10 group C, and 9/10 in group D. hNSE and HuNu staining revealed similar graft volume estimates in transplanted stroke animals. hNSE-immunoreactive fibers were also present within the corpus callosum, coursing in parallel with host tracts, suggesting a propensity to follow established neuroanatomical features. Despite absence of reduction in infarct volume, NSI-566RSC transplantation produced behavioral improvements possibly via robust engraftment and neuronal differentiation, supporting the use of this NSC line for stroke therapy.
Collapse
Affiliation(s)
- Naoki Tajiri
- Center of Excellence for Aging & Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa, Florida, United States of America
| | - David M. Quach
- Neuralstem, Inc., Rockville, Maryland, United States of America
| | - Yuji Kaneko
- Center of Excellence for Aging & Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa, Florida, United States of America
| | - Stephanie Wu
- Neuralstem, Inc., Rockville, Maryland, United States of America
| | - David Lee
- Neuralstem, Inc., Rockville, Maryland, United States of America
| | - Tina Lam
- Neuralstem, Inc., Rockville, Maryland, United States of America
| | - Ken L. Hayama
- Neuralstem, Inc., Rockville, Maryland, United States of America
| | - Thomas G. Hazel
- Neuralstem, Inc., Rockville, Maryland, United States of America
| | - Karl Johe
- Neuralstem, Inc., Rockville, Maryland, United States of America
| | - Michael C. Wu
- Neurodigitech, LLC., San Diego, California, United States of America
| | - Cesar V. Borlongan
- Center of Excellence for Aging & Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa, Florida, United States of America
- * E-mail:
| |
Collapse
|
50
|
Pin-Barre C, Laurin J, Felix MS, Pertici V, Kober F, Marqueste T, Matarazzo V, Muscatelli-Bossy F, Temprado JJ, Brisswalter J, Decherchi P. Acute neuromuscular adaptation at the spinal level following middle cerebral artery occlusion-reperfusion in the rat. PLoS One 2014; 9:e89953. [PMID: 24587147 PMCID: PMC3938604 DOI: 10.1371/journal.pone.0089953] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 01/24/2014] [Indexed: 11/18/2022] Open
Abstract
The purpose of the study was to highlight the acute motor reflex adaptation and to deepen functional deficits following a middle cerebral artery occlusion-reperfusion (MCAO-r). Thirty-six Sprague-Dawley rats were included in this study. The middle cerebral artery occlusion (MCAO; 120 min) was performed on 16 rats studied at 1 and 7 days, respectively (MCAO-D1 and MCAO-D7, n = 8 for each group). The other animals were divided into 3 groups: SHAM-D1 (n = 6), SHAM-D7 (n = 6) and Control (n = 8). Rats performed 4 behavioral tests (the elevated body swing test, the beam balance test, the ladder-climbing test and the forelimb grip force) before the surgery and daily after MCAO-r. H-reflex on triceps brachii was measured before and after isometric exercise. Infarction size and cerebral edema were respectively assessed by histological (Cresyl violet) and MRI measurements at the same time points than H-reflex recordings. Animals with cerebral ischemia showed persistent functional deficits during the first week post-MCAO-r. H-reflex was not decreased in response to isometric exercise one day after the cerebral ischemia contrary to the other groups. The motor reflex regulation was recovered 7 days post-MCAO-r. This result reflects an acute sensorimotor adaptation at the spinal level after MCAO-r.
Collapse
Affiliation(s)
- Caroline Pin-Barre
- Aix-Marseille Université, Centre National de la Recherche Scientifique, Institut des Sciences du Mouvement, Faculté des Sciences du Sport, Marseille, France
- Université de Nice Sophia-Antipolis et Université du Sud Toulon-Var, Motricité Humaine Éducation Sport Santé, Nice, France
| | - Jérôme Laurin
- Aix-Marseille Université, Centre National de la Recherche Scientifique, Institut des Sciences du Mouvement, Faculté des Sciences du Sport, Marseille, France
- * E-mail:
| | - Marie-Solenne Felix
- Aix-Marseille Université, Centre National de la Recherche Scientifique, Institut des Sciences du Mouvement, Faculté des Sciences du Sport, Marseille, France
| | - Vincent Pertici
- Aix-Marseille Université, Centre National de la Recherche Scientifique, Institut des Sciences du Mouvement, Faculté des Sciences du Sport, Marseille, France
| | - Frank Kober
- Aix-Marseille Université, Centre National de la Recherche Scientifique, Centre de Résonance Magnétique Biologique et Médicale, Faculté de Médecine Timone, Marseille, France
| | - Tanguy Marqueste
- Aix-Marseille Université, Centre National de la Recherche Scientifique, Institut des Sciences du Mouvement, Faculté des Sciences du Sport, Marseille, France
| | - Valery Matarazzo
- Aix-Marseille Université, Institut National de la Santé et de la Recherche Médicale, Institut de Neurobiologie de la Méditerranée, Marseille, France
| | - Françoise Muscatelli-Bossy
- Aix-Marseille Université, Institut National de la Santé et de la Recherche Médicale, Institut de Neurobiologie de la Méditerranée, Marseille, France
| | - Jean-Jacques Temprado
- Aix-Marseille Université, Centre National de la Recherche Scientifique, Institut des Sciences du Mouvement, Faculté des Sciences du Sport, Marseille, France
| | - Jeanick Brisswalter
- Université de Nice Sophia-Antipolis et Université du Sud Toulon-Var, Motricité Humaine Éducation Sport Santé, Nice, France
| | - Patrick Decherchi
- Aix-Marseille Université, Centre National de la Recherche Scientifique, Institut des Sciences du Mouvement, Faculté des Sciences du Sport, Marseille, France
| |
Collapse
|