1
|
Klein B, Ciesielska A, Losada PM, Sato A, Shah-Morales S, Ford JB, Higashikubo B, Tager D, Urry A, Bombosch J, Chang WC, Andrews-Zwilling Y, Nejadnik B, Warraich Z, Paz JT. Modified human mesenchymal stromal/stem cells restore cortical excitability after focal ischemic stroke in rats. Mol Ther 2025; 33:375-400. [PMID: 39668560 PMCID: PMC11764858 DOI: 10.1016/j.ymthe.2024.12.006] [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: 04/04/2024] [Revised: 09/18/2024] [Accepted: 12/06/2024] [Indexed: 12/14/2024] Open
Abstract
Allogeneic modified bone marrow-derived human mesenchymal stromal/stem cells (hMSC-SB623 cells) are in clinical development for the treatment of chronic motor deficits after traumatic brain injury and cerebral ischemic stroke. However, their exact mechanisms of action remain unclear. Here, we investigated the effects of this cell therapy on cortical network excitability, brain tissue, and peripheral blood at a chronic stage after ischemic stroke in a rat model. One month after focal cortical ischemic stroke, hMSC-SB623 cells or the vehicle solution were injected into the peri-stroke cortex. Starting one week after treatment, cortical excitability was assessed ex vivo. hMSC-SB623 cell transplants reduced stroke-induced cortical hyperexcitability, restoring cortical excitability to control levels. The histology of brain tissue revealed an increase of factors relevant to neuroregeneration, and synaptic and cellular plasticity. Whole-blood RNA sequencing and serum protein analyses showed that intra-cortical hMSC-SB623 cell transplantation reversed effects of stroke on peripheral blood factors known to be involved in stroke pathophysiology. Our findings demonstrate that intra-cortical transplants of hMSC-SB623 cells correct stroke-induced circuit disruptions even at the chronic stage, suggesting broad usefulness as a therapeutic for neurological conditions with network hyperexcitability. Additionally, the transplanted cells exert far-reaching immunomodulatory effects whose therapeutic impact remains to be explored.
Collapse
Affiliation(s)
| | - Agnieszka Ciesielska
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA; University of California, San Francisco, Department of Neurology, and the Kavli Institute for Fundamental Neuroscience, San Francisco, CA, USA
| | | | | | | | - Jeremy B Ford
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | | | - Dale Tager
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Alexander Urry
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | | | | | | | | | | | - Jeanne T Paz
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA; University of California, San Francisco, Department of Neurology, and the Kavli Institute for Fundamental Neuroscience, San Francisco, CA, USA; University of California, San Francisco, Neurosciences Graduate Program, San Francisco, CA, USA.
| |
Collapse
|
2
|
Goto A, Moriya Y, Nakayama M, Iwasaki S, Yamamoto S. DMPK perspective on quantitative model analysis for chimeric antigen receptor cell therapy: Advances and challenges. Drug Metab Pharmacokinet 2024; 56:101003. [PMID: 38843652 DOI: 10.1016/j.dmpk.2024.101003] [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: 11/01/2023] [Revised: 01/26/2024] [Accepted: 02/10/2024] [Indexed: 06/24/2024]
Abstract
Chimeric antigen receptor (CAR) cells are genetically engineered immune cells that specifically target tumor-associated antigens and have revolutionized cancer treatment, particularly in hematological malignancies, with ongoing investigations into their potential applications in solid tumors. This review provides a comprehensive overview of the current status and challenges in drug metabolism and pharmacokinetics (DMPK) for CAR cell therapy, specifically emphasizing on quantitative modeling and simulation (M&S). Furthermore, the recent advances in quantitative model analysis have been reviewed, ranging from clinical data characterization to mechanism-based modeling that connects in vitro and in vivo nonclinical and clinical study data. Additionally, the future perspectives and areas for improvement in CAR cell therapy translation have been reviewed. This includes using formulation quality considerations, characterization of appropriate animal models, refinement of in vitro models for bottom-up approaches, and enhancement of quantitative bioanalytical methodology. Addressing these challenges within a DMPK framework is pivotal in facilitating the translation of CAR cell therapy, ultimately enhancing the patients' lives through efficient CAR cell therapies.
Collapse
Affiliation(s)
- Akihiko Goto
- Center of Excellence for Drug Metabolism, Pharmacokinetics and Modeling, Preclinical and Translational Sciences, Research, Takeda Pharmaceutical Company Limited, Kanagawa, Japan
| | - Yuu Moriya
- Center of Excellence for Drug Metabolism, Pharmacokinetics and Modeling, Preclinical and Translational Sciences, Research, Takeda Pharmaceutical Company Limited, Kanagawa, Japan
| | - Miyu Nakayama
- Center of Excellence for Drug Metabolism, Pharmacokinetics and Modeling, Preclinical and Translational Sciences, Research, Takeda Pharmaceutical Company Limited, Kanagawa, Japan
| | - Shinji Iwasaki
- Center of Excellence for Drug Metabolism, Pharmacokinetics and Modeling, Preclinical and Translational Sciences, Research, Takeda Pharmaceutical Company Limited, Kanagawa, Japan
| | - Syunsuke Yamamoto
- Center of Excellence for Drug Metabolism, Pharmacokinetics and Modeling, Preclinical and Translational Sciences, Research, Takeda Pharmaceutical Company Limited, Kanagawa, Japan.
| |
Collapse
|
3
|
Kawabori M, Kuroda S, Shichinohe H, Kahata K, Shiratori S, Ikeda S, Harada T, Hirata K, Tha KK, Aragaki M, Terasaka S, Ito YM, Nishimoto N, Ohnishi S, Yabe I, Kudo K, Houkin K, Fujimura M. Intracerebral transplantation of MRI-trackable autologous bone marrow stromal cells for patients with subacute ischemic stroke. MED 2024; 5:432-444.e4. [PMID: 38547868 DOI: 10.1016/j.medj.2024.02.009] [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: 07/05/2023] [Revised: 10/19/2023] [Accepted: 02/26/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND Ischemic stroke is one of the leading causes of death and neurological disability worldwide, and stem cell therapy is highly expected to reverse the sequelae. This phase 1/2, first-in-human study evaluated the safety, feasibility, and monitoring of an intracerebral-transplanted magnetic resonance imaging (MRI)-trackable autologous bone marrow stromal cell (HUNS001-01) for patients with subacute ischemic stroke. METHODS The study included adults with severe disability due to ischemic stroke. HUNS001-01 cultured with human platelet lysates and labeled with superparamagnetic iron oxide was stereotactically transplanted into the peri-infarct area 47-64 days after ischemic stroke onset (dose: 2 or 5 × 107 cells). Neurological and radiographic evaluations were performed throughout 1 year after cell transplantation. The trial was registered at UMIN Clinical Trial Registry (number UMIN000026130). FINDINGS All seven patients who met the inclusion criteria successfully achieved cell expansion, underwent intracerebral transplantation, and completed 1 year of follow-up. No product-related adverse events were observed. The median National Institutes of Health Stroke Scale and modified Rankin scale scores before transplantation were 13 and 4, which showed improvements of 1-8 and 0-2, respectively. Cell tracking proved that the engrafted cells migrated toward the infarction border area 1-6 months after transplantation, and the quantitative susceptibility mapping revealed that cell signals at the migrated area constantly increased throughout the follow-up period up to 34% of that of the initial transplanted site. CONCLUSIONS Intracerebral transplantation of HUNS001-01 was safe and well tolerated. Cell tracking shed light on the therapeutic mechanisms of intracerebral transplantation. FUNDING This work was supported by the Japan Agency for Medical Research and Development (AMED; JP17bk0104045 and JP20bk0104011).
Collapse
Affiliation(s)
- Masahito Kawabori
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido 060-8638, Japan.
| | - Satoshi Kuroda
- Department of Neurosurgery, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Hideo Shichinohe
- Institute of Health Science Innovation for Medical Care, Hokkaido University Hospital, Sapporo, Hokkaido 060-8638, Japan
| | - Kaoru Kahata
- Institute of Health Science Innovation for Medical Care, Hokkaido University Hospital, Sapporo, Hokkaido 060-8638, Japan
| | - Souichi Shiratori
- Department of Hematology, Hokkaido University Faculty of Medicine, Sapporo, Hokkaido 060-8638, Japan
| | - Satoshi Ikeda
- Department of Rehabilitation, Hokkaido University Hospital, Sapporo, Hokkaido 060-8638, Japan
| | - Taisuke Harada
- Department of Diagnostic Imaging, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido 060-8638, Japan
| | - Kenji Hirata
- Department of Diagnostic Imaging, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido 060-8638, Japan
| | - Khin Khin Tha
- Global Center for Biomedical Science and Engineering, Hokkaido University Faculty of Medicine, Sapporo, Hokkaido 060-8638, Japan
| | - Masato Aragaki
- Institute of Health Science Innovation for Medical Care, Hokkaido University Hospital, Sapporo, Hokkaido 060-8638, Japan
| | - Shunsuke Terasaka
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido 060-8638, Japan
| | - Yoichi M Ito
- Institute of Health Science Innovation for Medical Care, Hokkaido University Hospital, Sapporo, Hokkaido 060-8638, Japan
| | - Naoki Nishimoto
- Institute of Health Science Innovation for Medical Care, Hokkaido University Hospital, Sapporo, Hokkaido 060-8638, Japan
| | - Shunsuke Ohnishi
- Laboratory of Molecular and Cellular Medicine, Hokkaido University Graduate School of Pharmacology, Sapporo, Hokkaido 060-8638, Japan
| | - Ichiro Yabe
- Department of Neurology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido 060-8638, Japan
| | - Kohsuke Kudo
- Department of Diagnostic Imaging, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido 060-8638, Japan
| | - Kiyohiro Houkin
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido 060-8638, Japan
| | - Miki Fujimura
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido 060-8638, Japan
| |
Collapse
|
4
|
Intracerebral Transplantation of Mesenchymal Stromal Cell Compounded with Recombinant Peptide Scaffold against Chronic Intracerebral Hemorrhage Model. Stem Cells Int 2022; 2022:8521922. [PMID: 35966129 PMCID: PMC9372516 DOI: 10.1155/2022/8521922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 06/17/2022] [Accepted: 06/28/2022] [Indexed: 11/18/2022] Open
Abstract
Background Due to the lack of effective therapies, stem cell transplantation is an anticipated treatment for chronic intracerebral hemorrhage (ICH), and higher cell survival and engraftment are considered to be the key for recovery. Mesenchymal stromal cells (MSCs) compounded with recombinant human collagen type I scaffolds (CellSaics) have a higher potential for cell survival and engraftment compared with solo-MSCs, and we investigated the validity of intracerebral transplantation of CellSaic in a chronic ICH model. Methods Rat CellSaics (rCellSaics) were produced by rat bone marrow-derived MSC (rBMSCs). The secretion potential of neurotrophic factors and the cell proliferation rate were compared under oxygen-glucose deprivation (OGD) conditions. rCellSaics, rBMSCs, or saline were transplanted into the hollow cavity of a rat chronic ICH model. Functional and histological analyses were evaluated, and single-photon emission computed tomography for benzodiazepine receptors was performed to monitor sequential changes in neuronal integrity. Furthermore, human CellSaics (hCellSaics) were transplanted into a chronic ICH model in immunodeficient rats. Antibodies neutralizing brain-derived neurotrophic factor (BDNF) were used to elucidate its mode of action. Results rCellSaics demonstrated a higher secretion potential of trophic factors and showed better cell proliferation in the OGD condition. Animals receiving rCellSaics displayed better neurological recovery, higher intracerebral BDNF, and better cell engraftment; they also showed a tendency for less brain atrophy and higher benzodiazepine receptor preservation. hCellSaics also promoted significant functional recovery, which was reversed by BDNF neutralization. Conclusion Intracerebral transplantation of CellSaics enabled neurological recovery in a chronic ICH model and may be a good option for clinical application.
Collapse
|
5
|
Szöllősi D, Hegedűs N, Veres DS, Futó I, Horváth I, Kovács N, Martinecz B, Dénes Á, Seifert D, Bergmann R, Lebeda O, Varga Z, Kaleta Z, Szigeti K, Máthé D. Evaluation of Brain Nuclear Medicine Imaging Tracers in a Murine Model of Sepsis-Associated Encephalopathy. Mol Imaging Biol 2019; 20:952-962. [PMID: 29736562 PMCID: PMC6244542 DOI: 10.1007/s11307-018-1201-3] [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] [Indexed: 12/13/2022]
Abstract
Purpose The purpose of this study was to evaluate a set of widely used nuclear medicine imaging agents as possible methods to study the early effects of systemic inflammation on the living brain in a mouse model of sepsis-associated encephalopathy (SAE). The lipopolysaccharide (LPS)-induced murine systemic inflammation model was selected as a model of SAE. Procedures C57BL/6 mice were used. A multimodal imaging protocol was carried out on each animal 4 h following the intravenous administration of LPS using the following tracers: [99mTc][2,2-dimethyl-3-[(3E)-3-oxidoiminobutan-2-yl]azanidylpropyl]-[(3E)-3-hydroxyiminobutan-2-yl]azanide ([99mTc]HMPAO) and ethyl-7-[125I]iodo-5-methyl-6-oxo-4H-imidazo[1,5-a][1,4]benzodiazepine-3-carboxylate ([125I]iomazenil) to measure brain perfusion and neuronal damage, respectively; 2-deoxy-2-[18F]fluoro-d-glucose ([18F]FDG) to measure cerebral glucose uptake. We assessed microglia activity on another group of mice using 2-[6-chloro-2-(4-[125I]iodophenyl)-imidazo[1,2-a]pyridin-3-yl]-N-ethyl-N-methyl-acetamide ([125I]CLINME). Radiotracer uptakes were measured in different brain regions and correlated. Microglia activity was also assessed using immunohistochemistry. Brain glutathione levels were measured to investigate oxidative stress. Results Significantly reduced perfusion values and significantly enhanced [18F]FDG and [125I]CLINME uptake was measured in the LPS-treated group. Following perfusion compensation, enhanced [125I]iomazenil uptake was measured in the LPS-treated group’s hippocampus and cerebellum. In this group, both [18F]FDG and [125I]iomazenil uptake showed highly negative correlation to perfusion measured with ([99mTc]HMPAO uptake in all brain regions. No significant differences were detected in brain glutathione levels between the groups. The CD45 and P2Y12 double-labeling immunohistochemistry showed widespread microglia activation in the LPS-treated group. Conclusions Our results suggest that [125I]CLINME and [99mTc]HMPAO SPECT can be used to detect microglia activation and brain hypoperfusion, respectively, in the early phase (4 h post injection) of systemic inflammation. We suspect that the enhancement of [18F]FDG and [125I]iomazenil uptake in the LPS-treated group does not necessarily reflect neural hypermetabolism and the lack of neuronal damage. They are most likely caused by processes emerging during neuroinflammation, e.g., microglia activation and/or immune cell infiltration. Electronic supplementary material The online version of this article (10.1007/s11307-018-1201-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Dávid Szöllősi
- Department of Biophysics and Radiation Biology, Semmelweis Univ, Budapest, H-1094, Hungary
| | - Nikolett Hegedűs
- Department of Biophysics and Radiation Biology, Semmelweis Univ, Budapest, H-1094, Hungary
| | - Dániel S Veres
- Department of Biophysics and Radiation Biology, Semmelweis Univ, Budapest, H-1094, Hungary
| | - Ildikó Futó
- Department of Biophysics and Radiation Biology, Semmelweis Univ, Budapest, H-1094, Hungary
| | - Ildikó Horváth
- Department of Biophysics and Radiation Biology, Semmelweis Univ, Budapest, H-1094, Hungary
| | - Noémi Kovács
- CROmed Translational Research Centers, Budapest, H-1047, Hungary
| | - Bernadett Martinecz
- Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Ádám Dénes
- Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Daniel Seifert
- Nuclear Physics Institute of the CAS, CZ 250 68, Rez, Czech Republic
| | - Ralf Bergmann
- Helmholz-Zentrum Dresden-Rossendorf, Radiopharmazie Radiopharmaceutische Biologie, Dresden, Germany
| | - Ondřej Lebeda
- Nuclear Physics Institute of the CAS, CZ 250 68, Rez, Czech Republic
| | - Zoltán Varga
- Department of Biophysics and Radiation Biology, Semmelweis Univ, Budapest, H-1094, Hungary.,Biological Nanochemistry Research Group, Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Zoltán Kaleta
- Progressio Fine Chemical Engineering Ltd, Székesfehérvár, Hungary
| | - Krisztián Szigeti
- Department of Biophysics and Radiation Biology, Semmelweis Univ, Budapest, H-1094, Hungary.
| | - Domokos Máthé
- CROmed Translational Research Centers, Budapest, H-1047, Hungary
| |
Collapse
|
6
|
Yun CW, Lee SH. Enhancement of Functionality and Therapeutic Efficacy of Cell-Based Therapy Using Mesenchymal Stem Cells for Cardiovascular Disease. Int J Mol Sci 2019; 20:ijms20040982. [PMID: 30813471 PMCID: PMC6412804 DOI: 10.3390/ijms20040982] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 02/20/2019] [Accepted: 02/21/2019] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular disease usually triggers coronary heart disease, stroke, and ischemic diseases, thus promoting the development of functional failure. Mesenchymal stem cells (MSCs) are cells that can be isolated from various human tissues, with multipotent and immunomodulatory characteristics to help damaged tissue repair and avoidance of immune responses. Much research has proved the feasibility, safety, and efficiency of MSC-based therapy for cardiovascular disease. Despite the fact that the precise mechanism of MSCs remains unclear, their therapeutic capability to treat ischemic diseases has been tested in phase I/II clinical trials. MSCs have the potential to become an effective therapeutic strategy for the treatment of ischemic and non-ischemic cardiovascular disorders. The molecular mechanism underlying the efficacy of MSCs in promoting engraftment and accelerating the functional recovery of injury sites is still unclear. It is hypothesized that the mechanisms of paracrine effects for the cardiac repair, optimization of the niche for cell survival, and cardiac remodeling by inflammatory control are involved in the interaction between MSCs and the damaged myocardial environment. This review focuses on recent experimental and clinical findings related to cardiovascular disease. We focus on MSCs, highlighting their roles in cardiovascular disease repair, differentiation, and MSC niche, and discuss their therapeutic efficacy and the current status of MSC-based cardiovascular disease therapies.
Collapse
Affiliation(s)
- Chul Won Yun
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul 04401, Korea.
| | - Sang Hun Lee
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul 04401, Korea.
- Department of Biochemistry, Soonchunhyang University College of Medicine, Cheonan 34538, Korea.
| |
Collapse
|
7
|
Tan C, Zhao S, Higashikawa K, Wang Z, Kawabori M, Abumiya T, Nakayama N, Kazumata K, Ukon N, Yasui H, Tamaki N, Kuge Y, Shichinohe H, Houkin K. [ 18F]DPA-714 PET imaging shows immunomodulatory effect of intravenous administration of bone marrow stromal cells after transient focal ischemia. EJNMMI Res 2018; 8:35. [PMID: 29717383 PMCID: PMC5930298 DOI: 10.1186/s13550-018-0392-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 04/23/2018] [Indexed: 12/22/2022] Open
Abstract
Background The potential application of bone marrow stromal cell (BMSC) therapy in stroke has been anticipated due to its immunomodulatory effects. Recently, positron emission tomography (PET) with [18F]DPA-714, a translocator protein (TSPO) ligand, has become available for use as a neural inflammatory indicator. We aimed to evaluate the effects of BMSC administration after transient middle cerebral artery occlusion (MCAO) using [18F]DPA-714 PET. The BMSCs or vehicle were administered intravenously to rat MCAO models at 3 h after the insult. Neurological deficits, body weight, infarct volume, and histology were analyzed. [18F]DPA-714 PET was performed 3 and 10 days after MCAO. Results Rats had severe neurological deficits and body weight loss after MCAO. Cell administration ameliorated these effects as well as the infarct volume. Although weight loss occurred in the spleen and thymus, cell administration suppressed it. In both vehicle and BMSC groups, [18F]DPA-714 PET showed a high standardized uptake value (SUV) around the ischemic area 3 days after MCAO. Although SUV was increased further 10 days after MCAO in both groups, the increase was inhibited in the BMSC group, significantly. Histological analysis showed that an inflammatory reaction occurred in the lymphoid organs and brain after MCAO, which was suppressed in the BMSC group. Conclusions The present results suggest that BMSC therapy could be effective in ischemic stroke due to modulation of systemic inflammatory responses. The [18F]DPA-714 PET/CT system can accurately demonstrate brain inflammation and evaluate the BMSC therapeutic effect in an imaging context. It has great potential for clinical application.
Collapse
Affiliation(s)
- Chengbo Tan
- Department of Neurosurgery, Graduate School of Medicine, Hokkaido University, N15 W7, Kita-ku, Sapporo, 060-8638, Japan.,Advanced Clinical Research Center, Fukushima Global Medical Science Center, Fukushima Medical University, Fukushima, Japan
| | - Songji Zhao
- Advanced Clinical Research Center, Fukushima Global Medical Science Center, Fukushima Medical University, Fukushima, Japan.,Department of Tracer Kinetics and Bioanalysis, Graduate School of Medicine, Hokkaido University, Sapporo, Japan.,Department of Nuclear Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Kei Higashikawa
- Central Institute of Isotope Science, Hokkaido University, Sapporo, Japan.,Department of Integrated Molecular Imaging, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Zifeng Wang
- Department of Neurosurgery, Graduate School of Medicine, Hokkaido University, N15 W7, Kita-ku, Sapporo, 060-8638, Japan
| | - Masahito Kawabori
- Department of Neurosurgery, Graduate School of Medicine, Hokkaido University, N15 W7, Kita-ku, Sapporo, 060-8638, Japan
| | - Takeo Abumiya
- Department of Neurosurgery, Graduate School of Medicine, Hokkaido University, N15 W7, Kita-ku, Sapporo, 060-8638, Japan
| | - Naoki Nakayama
- Department of Neurosurgery, Graduate School of Medicine, Hokkaido University, N15 W7, Kita-ku, Sapporo, 060-8638, Japan
| | - Ken Kazumata
- Department of Neurosurgery, Graduate School of Medicine, Hokkaido University, N15 W7, Kita-ku, Sapporo, 060-8638, Japan
| | - Naoyuki Ukon
- Advanced Clinical Research Center, Fukushima Global Medical Science Center, Fukushima Medical University, Fukushima, Japan.,Department of Nuclear Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan.,Central Institute of Isotope Science, Hokkaido University, Sapporo, Japan
| | - Hironobu Yasui
- Central Institute of Isotope Science, Hokkaido University, Sapporo, Japan.,Department of Integrated Molecular Imaging, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Nagara Tamaki
- Department of Nuclear Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yuji Kuge
- Central Institute of Isotope Science, Hokkaido University, Sapporo, Japan.,Department of Integrated Molecular Imaging, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hideo Shichinohe
- Department of Neurosurgery, Graduate School of Medicine, Hokkaido University, N15 W7, Kita-ku, Sapporo, 060-8638, Japan. .,Division of Clinical Research Administration, Hokkaido University Hospital, Sapporo, Japan.
| | - Kiyohiro Houkin
- Department of Neurosurgery, Graduate School of Medicine, Hokkaido University, N15 W7, Kita-ku, Sapporo, 060-8638, Japan
| |
Collapse
|
8
|
Shichinohe H, Kawabori M, Iijima H, Teramoto T, Abumiya T, Nakayama N, Kazumata K, Terasaka S, Arato T, Houkin K. Research on advanced intervention using novel bone marrOW stem cell (RAINBOW): a study protocol for a phase I, open-label, uncontrolled, dose-response trial of autologous bone marrow stromal cell transplantation in patients with acute ischemic stroke. BMC Neurol 2017; 17:179. [PMID: 28886699 PMCID: PMC5591569 DOI: 10.1186/s12883-017-0955-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 08/28/2017] [Indexed: 11/20/2022] Open
Abstract
Background Stroke is a leading cause of death and disability, and despite intensive research, few treatment options exist. However, a recent breakthrough in cell therapy is expected to reverse the neurological sequelae of stroke. Although some pioneer studies on the use of cell therapy for treating stroke have been reported, certain problems remain unsolved. Recent studies have demonstrated that bone marrow stromal cells (BMSCs) have therapeutic potential against stroke. We investigated the use of autologous BMSC transplantation as a next-generation cell therapy for treating stroke. In this article, we introduce the protocol of a new clinical trial, the Research on Advanced Intervention using Novel Bone marrOW stem cell (RAINBOW). Methods/design RAINBOW is a phase 1, open-label, uncontrolled, dose-response study, with the primary aim to determine the safety of the autologous BMSC product HUNS001–01 when administered to patients with acute ischemic stroke. Estimated enrollment is 6–10 patients suffering from moderate to severe neurological deficits. Approximately 50 mL of the bone marrow is extracted from the iliac bone of each patient 15 days or later from the onset. BMSCs are cultured with allogeneic human platelet lysate (PL) as a substitute for fetal calf serum and are labeled with superparamagnetic iron oxide for cell tracking using magnetic resonance imaging (MRI). HUNS001–01 is stereotactically administered around the area of infarction in the subacute phase. Each patient will be administered a dose of 20 or 50 million cells. Neurological scoring, MRI for cell tracking, 18F–fuorodeoxyglucose positron emission tomography, and 123I–Iomazenil singlephoton emission computed tomography will be performed for 1 year after the administration. Discussion This is a first-in-human trial for HUNS001–01 to the patients with acute ischemic stroke. We expect that intraparenchymal injection can be a more favorable method for cell delivery to the lesion and improvement of the motor function than intravenous infusion. Moreover, it is expected that the bio-imaging techniques can clarify the therapeutic mechanisms. Trial registration The trial was registered at The University Hospital Medical Information Network on February 22, 2017 (UNIN ID: UMIN000026130). The findings of this trial will be disseminated to patients and through peer-reviewed publications and international presentations.
Collapse
Affiliation(s)
- Hideo Shichinohe
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan. .,Clinical Research and Medical Innovation Center, Hokkaido University Hospital, N14 W5, Kita-ku, Sapporo, 060-8648, Japan.
| | - Masahito Kawabori
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Hiroaki Iijima
- Clinical Research and Medical Innovation Center, Hokkaido University Hospital, N14 W5, Kita-ku, Sapporo, 060-8648, Japan
| | - Tuyoshi Teramoto
- Clinical Research and Medical Innovation Center, Hokkaido University Hospital, N14 W5, Kita-ku, Sapporo, 060-8648, Japan
| | - Takeo Abumiya
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Naoki Nakayama
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Ken Kazumata
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Shunsuke Terasaka
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Teruyo Arato
- Clinical Research and Medical Innovation Center, Hokkaido University Hospital, N14 W5, Kita-ku, Sapporo, 060-8648, Japan
| | - Kiyohiro Houkin
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| |
Collapse
|
9
|
Long Q, Luo Q, Wang K, Bates A, Shetty AK. Mash1-dependent Notch Signaling Pathway Regulates GABAergic Neuron-Like Differentiation from Bone Marrow-Derived Mesenchymal Stem Cells. Aging Dis 2017; 8:301-313. [PMID: 28580186 PMCID: PMC5440110 DOI: 10.14336/ad.2016.1018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 10/18/2016] [Indexed: 12/16/2022] Open
Abstract
GABAergic neuronal cell grafting has promise for treating a multitude of neurological disorders including epilepsy, age-related memory dysfunction, Alzheimer's disease and schizophrenia. However, identification of an unlimited source of GABAergic cells is critical for advancing such therapies. Our previous study implied that reprogramming of bone marrow-derived mesenchymal stem cells (BMSCs) through overexpression of the Achaete-scute homolog 1 (Ascl1, also called Mash1) could generate GABAergic neuron-like cells. Here, we investigated mechanisms underlying the conversion of BMSCs into GABAergic cells. We inhibited γ-secretase (an enzyme that activates Notch signaling) with N-[N-(3,5-difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester (DAPT) or manipulated the expression of Notch signaling components such as the recombination signal binding protein for immunoglobulin kappa J region (RBPJ), hairy and enhancer of split-1 (Hes1) or Mash1. We demonstrate that inhibition of γ-secretase through DAPT down-regulates RBPJ and Hes1, up-regulates Mash1 and results in an enhanced differentiation of BMSCs into GABAergic cells. On the other hand, RBPJ knockdown in BMSCs has no effect on Mash1 gene expression whereas Hes1 knockdown increases the expression of Mash1. Transduction of Mash1 in BMSCs also increases the expression of Hes1 but not RBPJ. Moreover, increased GABAergic differentiation in BMSCs occurs with concurrent Mash1 overexpression and Hes1-silencing. Thus, the Mash1-dependent Notch signaling pathway regulates GABAergic neuron-like differentiation of BMSCs. These results also suggest that genetic engineering of BMSCs is a useful avenue for obtaining GABAergic neuron-like donor cells for the treatment of neurological disorders.
Collapse
Affiliation(s)
- Qianfa Long
- 1Department of Neurosurgery, Xi'an Central Hospital, Xi'an Jiao Tong University School of Medicine, Xi'an 710003, China.,2Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine at Scott & White, Temple and College Station, Texas, 76502, USA
| | - Qiang Luo
- 1Department of Neurosurgery, Xi'an Central Hospital, Xi'an Jiao Tong University School of Medicine, Xi'an 710003, China
| | - Kai Wang
- 3Department of Neurosurgery, Qingdao 401 Hospital of PLA, Qingdao 266071, China
| | - Adrian Bates
- 2Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine at Scott & White, Temple and College Station, Texas, 76502, USA.,4Research Service, Olin E. Teague Veterans' Medical Center, CTVHCS, Temple, Texas, USA
| | - Ashok K Shetty
- 2Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine at Scott & White, Temple and College Station, Texas, 76502, USA.,4Research Service, Olin E. Teague Veterans' Medical Center, CTVHCS, Temple, Texas, USA
| |
Collapse
|
10
|
Kurisu K, Abumiya T, Ito M, Gekka M, Osanai T, Shichinohe H, Nakayama N, Kazumata K, Houkin K. Transarterial regional hypothermia provides robust neuroprotection in a rat model of permanent middle cerebral artery occlusion with transient collateral hypoperfusion. Brain Res 2016; 1651:95-103. [PMID: 27663968 DOI: 10.1016/j.brainres.2016.09.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 08/31/2016] [Accepted: 09/12/2016] [Indexed: 11/20/2022]
Abstract
The robust neuroprotective effects of transarterial regional hypothermia have been demonstrated in the typical transient middle cerebral artery occlusion (tMCAO) model, but have not yet been tested in other ischemic stroke models, even though clinical ischemic conditions are diverse. In order to clarify these effects in a different ischemic stroke model, we employed a rat model of permanent MCAO (pMCAO) with transient collateral hypoperfusion (tCHP), which was achieved by direct MCA ligation through craniotomy and 1-h bilateral common carotid artery occlusion at the beginning of pMCAO. The infusion of 20ml/kg of 4°C cold saline (CS) or 37°C warm saline (WS) into the ipsilateral internal carotid artery (ICA) was performed for 15min in intra- or post-tCHP. Neurological scores, infarct/edema volumes, and neuronal apoptosis and reactive gliosis were compared between the CS and WS groups and a non-infusion control group after 48h of reperfusion. Although brain temperatures were only reduced by 2-3°C for 15min, the CS group had significantly better neurological scores, smaller infarct/edema volumes, and less penumbral neuronal apoptosis and reactive gliosis than the control and WS groups. The post-tCHP CS group exhibited prominent neuroprotective effects, even though infarct volumes and neuronal apoptosis were reduced less than those in the intra-tCHP CS group. In conclusion, we demonstrated the neuroprotective effects of transarterial regional hypothermia in an ischemic model of pMCAO with tCHP. Even though MCAO is persistent, cold infusion via the ICA is neuroprotective for the penumbra, suggesting the wider therapeutic application of this therapy.
Collapse
Affiliation(s)
- Kota Kurisu
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido 060-8638, Japan
| | - Takeo Abumiya
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido 060-8638, Japan.
| | - Masaki Ito
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido 060-8638, Japan
| | - Masayuki Gekka
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido 060-8638, Japan
| | - Toshiya Osanai
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido 060-8638, Japan
| | - Hideo Shichinohe
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido 060-8638, Japan
| | - Naoki Nakayama
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido 060-8638, Japan
| | - Ken Kazumata
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido 060-8638, Japan
| | - Kiyohiro Houkin
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido 060-8638, Japan
| |
Collapse
|
11
|
Shichinohe H, Houkin K. Cell Therapy for Stroke: Review of Previous Clinical Trials and Introduction of Our New Trials. Neurol Med Chir (Tokyo) 2016; 56:592-596. [PMID: 27302193 PMCID: PMC5066079 DOI: 10.2176/nmc.st.2016-0087] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Stroke is still a leading cause of death and disability, and despite intensive research, few treatment options exist. A recent breakthrough in cell therapy is expected to reverse the neurological sequelae of stroke. Although some pioneer studies on the use of cell therapy for the treatment of stroke have been reported, certain problems still remain unsolved. We investigated the use of autologous bone marrow stromal cell (BMSC) transplantation for the treatment of stroke, to develop it as the next-generation cell therapy. In this study, we introduce the preparation of a new clinical trial, the Research on Advanced Intervention using Novel Bone marrow stem cell (RAINBOW) study. The trial will start in 2016, and we hope that it will not only be helpful for treating patients but also for clarifying the therapeutic mechanisms. Moreover, we review stem cell therapeutics as an emerging paradigm in stroke (STEPS) and the guidelines for the development of cell therapy for stroke in the United States as well as introduce the development of new guidelines in Japan. These guidelines are expected to encourage the development of cell therapy for stroke management.
Collapse
Affiliation(s)
- Hideo Shichinohe
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine
| | | |
Collapse
|
12
|
Kuroda S. Current Opinion of Bone Marrow Stromal Cell Transplantation for Ischemic Stroke. Neurol Med Chir (Tokyo) 2016; 56:293-301. [PMID: 26984453 PMCID: PMC4908072 DOI: 10.2176/nmc.ra.2015-0349] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
This article reviews recent advancement and perspective of bone marrow stromal cell (BMSC) transplantation for ischemic stroke, based on current information of basic and translational research. The author would like to emphasize that scientific approach would enable us to apply BMSC transplantation into clinical situation in near future.
Collapse
Affiliation(s)
- Satoshi Kuroda
- Department of Neurosurgery, Graduate School of Medicine and Pharmaceutical Science, University of Toyama
| |
Collapse
|
13
|
Suda S, Yang B, Schaar K, Xi X, Pido J, Parsha K, Aronowski J, Savitz SI. Autologous Bone Marrow Mononuclear Cells Exert Broad Effects on Short- and Long-Term Biological and Functional Outcomes in Rodents with Intracerebral Hemorrhage. Stem Cells Dev 2015; 24:2756-66. [PMID: 26414707 DOI: 10.1089/scd.2015.0107] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Autologous bone marrow-derived mononuclear cells (MNCs) are a potential therapy for ischemic stroke. However, the effect of MNCs in intracerebral hemorrhage (ICH) has not been fully studied. In this study, we investigated the effects of autologous MNCs in experimental ICH. ICH was induced by infusion of autologous blood into the left striatum in young and aged male Long Evans rats. Twenty-four hours after ICH, rats were randomized to receive an intravenous administration of autologous MNCs (1 × 10(7) cells/kg) or saline. We examined brain water content, various markers related to the integrity of the neurovascular unit and inflammation, neurological deficit, neuroregeneration, and brain atrophy. We found that MNC-treated young rats showed a reduction in the neurotrophil infiltration, the number of inducible nitric oxide synthase-positive cells, and the expression of inflammatory-related signalings such as the high-mobility group protein box-1, S100 calcium binding protein B, matrix metalloproteinase-9, and aquaporin 4. Ultimately, MNCs reduced brain edema in the perihematomal area compared with saline-treated animals at 3 days after ICH. Moreover, MNCs increased vessel density and migration of doublecortin-positive cells, improved motor functional recovery, spatial learning, and memory impairment, and reduced brain atrophy compared with saline-treated animals at 28 days after ICH. We also found that MNCs reduced brain edema and brain atrophy and improved spatial learning and memory in aged rats after ICH. We conclude that autologous MNCs can be safely harvested and intravenously reinfused in rodent ICH and may improve long-term structural and functional recovery after ICH. The results of this study may be applicable when considering future clinical trials testing MNCs for ICH.
Collapse
Affiliation(s)
- Satoshi Suda
- 1 Department of Neurological Science, Graduate School of Medicine, Nippon Medical School , Tokyo, Japan
| | - Bing Yang
- 2 Department of Neurology, University of Texas Medical School at Houston , Houston, Texas
| | - Krystal Schaar
- 2 Department of Neurology, University of Texas Medical School at Houston , Houston, Texas
| | - Xiaopei Xi
- 2 Department of Neurology, University of Texas Medical School at Houston , Houston, Texas
| | - Jennifer Pido
- 2 Department of Neurology, University of Texas Medical School at Houston , Houston, Texas
| | - Kaushik Parsha
- 2 Department of Neurology, University of Texas Medical School at Houston , Houston, Texas
| | - Jaroslaw Aronowski
- 2 Department of Neurology, University of Texas Medical School at Houston , Houston, Texas
| | - Sean I Savitz
- 2 Department of Neurology, University of Texas Medical School at Houston , Houston, Texas
| |
Collapse
|
14
|
Buyang Huanwu Decoction (BYHWD) Enhances Angiogenic Effect of Mesenchymal Stem Cell by Upregulating VEGF Expression After Focal Cerebral Ischemia. J Mol Neurosci 2015; 56:898-906. [PMID: 25796380 DOI: 10.1007/s12031-015-0539-0] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 03/04/2015] [Indexed: 12/28/2022]
Abstract
Buyang Huanwu decoction (BYHWD) has been used for centuries to treat paralysis and stroke. Previously, we have demonstrated that BYHWD combined with mesenchymal stem cell (MSC) transplantation attenuates ischemic injury partly by upregulating angiogenesis. However, the mechanisms of this drug for stroke treatment are not completely understood. Here, we aimed to clarify the mechanism of BYHWD on angiogenesis mediated by MSCs. Firstly, we verified microvessels with a size of 50-100 nm produced by either MSCs or MSCs treated by 500 μg/ml BYHWD. These exosomes were purified and found to be able to activate vascular endothelial growth factor (VEGF) expression in endothelial cells (ECs). Moreover, exosomes from MSCs and MSCs treated by BYHWD induced elevated microRNA (miRNA)-126 expression and reduced miR-221 and miR-222 expression. In MSCs, disruption of dicer, an enzyme responsible for miRNA maturation, by dicer small interfering RNA (siRNA), or RNase pretreatment abolished this ability of the exosomes. Additionally, exosomes from MSCs treated by BYHWD promoted VEGF and Ki-67 expression and augmented vascular density in rat brain after bilateral carotid artery ligation. In conclusion, our study revealed that BYHWD exposure augmented angiogenetic miRNA and VEGF expression in exosomes secreted by MSCs and elevated angiogenesis in rat brain.
Collapse
|
15
|
Ikegame Y, Yamashita K, Nakashima S, Nomura Y, Yonezawa S, Asano Y, Shinoda J, Hara H, Iwama T. Fate of graft cells: what should be clarified for development of mesenchymal stem cell therapy for ischemic stroke? Front Cell Neurosci 2014; 8:322. [PMID: 25374506 PMCID: PMC4204523 DOI: 10.3389/fncel.2014.00322] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Accepted: 09/24/2014] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are believed to be promising for cell administration therapy after ischemic stroke. Because of their advantageous characteristics, such as ability of differentiation into neurovascular lineages, avoidance of immunological problems, and abundance of graft cells in mesodermal tissues, studies regarding MSC therapy have increased recently. However, several controversies are yet to be resolved before a worldwide consensus regarding a standard protocol is obtained. In particular, the neuroprotective effects, the rate of cell migration to the lesion, and differentiation direction differ depending on preclinical observations. Analyses of these differences and application of recent developments in stem cell biology or engineering in imaging modality may contribute to identification of criteria for optimal stem cell therapy in which reliable protocols, which control cell quality and include safe administration procedures, are defined for each recovery phase after cerebral ischemia. In this mini review, we examine controversies regarding the fate of grafts and the prospects for advanced therapy that could be obtained through recent developments in stem cell research as direct conversion to neural cells.
Collapse
Affiliation(s)
- Yuka Ikegame
- Department of Neurosurgery, Chubu Medical Center for Prolonged Traumatic Brain Dysfunction Gifu, Japan ; Department of Clinical Brain Sciences, Gifu University Graduate School of Medicine Gifu, Japan ; Department of Cell Signaling, Gifu University Graduate School of Medicine Gifu, Japan
| | - Kentaro Yamashita
- Department of Neurosurgery, Gifu University Graduate School of Medicine Gifu, Japan ; Department of Neurosurgery, Murakami Memorial Hospital, Asahi University Gifu, Japan
| | - Shigeru Nakashima
- Department of Cell Signaling, Gifu University Graduate School of Medicine Gifu, Japan
| | - Yuichi Nomura
- Department of Neurosurgery, Chubu Medical Center for Prolonged Traumatic Brain Dysfunction Gifu, Japan
| | - Shingo Yonezawa
- Department of Neurosurgery, Chubu Medical Center for Prolonged Traumatic Brain Dysfunction Gifu, Japan
| | - Yoshitaka Asano
- Department of Neurosurgery, Chubu Medical Center for Prolonged Traumatic Brain Dysfunction Gifu, Japan ; Department of Clinical Brain Sciences, Gifu University Graduate School of Medicine Gifu, Japan
| | - Jun Shinoda
- Department of Neurosurgery, Chubu Medical Center for Prolonged Traumatic Brain Dysfunction Gifu, Japan ; Department of Clinical Brain Sciences, Gifu University Graduate School of Medicine Gifu, Japan
| | - Hideaki Hara
- Department of Biofunctional Evaluation, Molecular Pharmacology, Gifu Pharmaceutical University Gifu, Japan
| | - Toru Iwama
- Department of Neurosurgery, Gifu University Graduate School of Medicine Gifu, Japan
| |
Collapse
|