1
|
Liu R, Berry R, Wang L, Chaudhari K, Winters A, Sun Y, Caballero C, Ampofo H, Shi Y, Thata B, Colon-Perez L, Sumien N, Yang SH. Experimental Ischemic Stroke Induces Secondary Bihemispheric White Matter Degeneration and Long-Term Cognitive Impairment. Transl Stroke Res 2024:10.1007/s12975-024-01241-0. [PMID: 38488999 DOI: 10.1007/s12975-024-01241-0] [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: 12/18/2023] [Revised: 02/22/2024] [Accepted: 03/08/2024] [Indexed: 03/17/2024]
Abstract
Clinical studies have identified widespread white matter degeneration in ischemic stroke patients. However, contemporary research in stroke has predominately focused on the infarct and periinfarct penumbra regions. The involvement of white matter degeneration after ischemic stroke and its contribution to post-stroke cognitive impairment and dementia (PSCID) has remained less explored in experimental models. In this study, we examined the progression of locomotor and cognitive function up to 4 months after inducing ischemic stroke by middle cerebral artery occlusion in young adult rats. Despite evident ongoing locomotor recovery, long-term cognitive and affective impairments persisted after ischemic stroke, as indicated by Morris water maze, elevated plus maze, and open field performance. At 4 months after stroke, multimodal MRI was conducted to assess white matter degeneration. T2-weighted MRI (T2WI) unveiled bilateral cerebroventricular enlargement after ischemic stroke. Fluid Attenuated Inversion Recovery MRI (FLAIR) revealed white matter hyperintensities in the corpus callosum and fornix across bilateral hemispheres. A positive association between the volume of white matter hyperintensities and total cerebroventricular volume was noted in stroke rats. Further evidence of bilateral white matter degeneration was indicated by the reduction of fractional anisotropy and quantitative anisotropy at bilateral corpus callosum in diffusion-weighted MRI (DWI) analysis. Additionally, microglia and astrocyte activation were identified in the bilateral corpus callosum after stroke. Our study suggests that experimental ischemic stroke induced by MCAO in young rat replicate long-term cognitive impairment and bihemispheric white matter degeneration observed in ischemic stroke patients. This model provides an invaluable tool for unraveling the mechanisms underlying post-stroke secondary white matter degeneration and its contribution to PSCID.
Collapse
Affiliation(s)
- Ran Liu
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA
| | - Raymond Berry
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA
| | - Linshu Wang
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA
| | - Kiran Chaudhari
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA
| | - Ali Winters
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA
| | - Yuanhong Sun
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA
| | - Claire Caballero
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA
| | - Hannah Ampofo
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA
| | - Yiwei Shi
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA
| | - Bibek Thata
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA
| | - Luis Colon-Perez
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA
| | - Nathalie Sumien
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA
| | - Shao-Hua Yang
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA.
| |
Collapse
|
2
|
Yamashita T, Sasaki M, Sasaki Y, Nagahama H, Oka S, Kataoka-Sasaki Y, Ukai R, Yokoyama T, Kobayashi M, Kakizawa M, Kocsis JD, Honmou O. Rehabilitation facilitates functional improvement following intravenous infusion of mesenchymal stem cells in the chronic phase of cerebral ischemia in rats. Brain Res 2024; 1825:148709. [PMID: 38072373 DOI: 10.1016/j.brainres.2023.148709] [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: 10/29/2023] [Revised: 12/04/2023] [Accepted: 12/05/2023] [Indexed: 12/18/2023]
Abstract
The primary objective of this study was to investigate the potential facilitating effects of daily rehabilitation for chronic cerebral ischemia following the intravenous infusion of mesenchymal stem cells (MSC) in rats. The middle cerebral artery (MCA) was occluded by intraluminal occlusion using a microfilament (MCAO). Eight weeks after MCAO induction, the rats were used as a chronic cerebral ischemia model. Four experimental groups were studied: Vehicle group (medium only, no cells); Rehab group (vehicle + rehabilitation), MSC group (MSC only); and Combined group (MSC + rehabilitation). Rat MSCs were intravenously infused eight weeks after MCAO induction, and the rats received daily rehabilitation through treadmill exercise for 20 min. Behavioral testing, lesion volume assessment using magnetic resonance imaging (MRI), and histological analysis were performed during the observation period until 16 weeks after MCAO induction. All treated animals showed functional improvement compared with the Vehicle group; however, the therapeutic efficacy was greatest in the Combined group. The combination therapy is associated with enhanced neural plasticity shown with histological analysis and MRI diffusion tensor imaging. These findings provide behavioral evidence for enhanced recovery by combined therapy with rehabilitation and intravenous infusion of MSCs, and may form the basis for the development of clinical protocols in the future.
Collapse
Affiliation(s)
- Tatsuro Yamashita
- Department of Neural Regenerative Medicine, Institute of Regenerative Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan; Department of Rehabilitation, Sapporo Medical University School of Medicine, Sapporo, Hokkaido, Japan
| | - Masanori Sasaki
- Department of Neural Regenerative Medicine, Institute of Regenerative Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan; Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA.
| | - Yuichi Sasaki
- Department of Neural Regenerative Medicine, Institute of Regenerative Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan; Department of Rehabilitation, Sapporo Medical University School of Medicine, Sapporo, Hokkaido, Japan
| | - Hiroshi Nagahama
- Department of Neural Regenerative Medicine, Institute of Regenerative Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan; Division of Radioisotope Research, Biomedical Research, Education and Instrumentation Center, Sapporo Medical University School of Medicine, Sapporo, Hokkaido 060-8556, Japan
| | - Shinichi Oka
- Department of Neural Regenerative Medicine, Institute of Regenerative Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Yuko Kataoka-Sasaki
- Department of Neural Regenerative Medicine, Institute of Regenerative Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Ryo Ukai
- Department of Neural Regenerative Medicine, Institute of Regenerative Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Takahiro Yokoyama
- Department of Neural Regenerative Medicine, Institute of Regenerative Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Masato Kobayashi
- Department of Neural Regenerative Medicine, Institute of Regenerative Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan; Department of Rehabilitation, Sapporo Medical University School of Medicine, Sapporo, Hokkaido, Japan
| | - Masafumi Kakizawa
- Department of Rehabilitation, Sapporo Medical University School of Medicine, Sapporo, Hokkaido, Japan
| | - Jeffery D Kocsis
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA; Department of Neuroscience, Yale University School of Medicine, Neurology, PO BOX 208018, New Haven, CT 06510, USA; Center for Neuroscience and Regeneration Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Osamu Honmou
- Department of Neural Regenerative Medicine, Institute of Regenerative Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan; Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
3
|
Kurihara K, Sasaki M, Nagahama H, Obara H, Fukushi R, Hirota R, Yoshimoto M, Teramoto A, Kocsis JD, Yamashita T, Honmou O. Repeated intravenous infusion of mesenchymal stem cells enhances recovery of motor function in a rat model with chronic spinal cord injury. Brain Res 2023; 1817:148484. [PMID: 37442249 DOI: 10.1016/j.brainres.2023.148484] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/05/2023] [Accepted: 07/10/2023] [Indexed: 07/15/2023]
Abstract
Spinal cord injury (SCI) can cause paralysis with a high disease burden with limited treatment options. A single intravenous infusion of mesenchymal stem cells (MSCs) improves motor function in rat SCI models, possibly through the induction of axonal sprouting and remyelination. Repeated infusions (thrice at weekly intervals) of MSCs were administered to rats with chronic SCI to determine if multiple-dosing regimens enhance motor improvement. Chronic SCI rats were randomized and infused with vehicle (vehicle), single MSC injection at week 6 (MSC-1) or repeatedly injections of MSCs at 6, 7, and 8 weeks (MSC-3) after SCI induction. In addition, a single high dose of MSCs (HD-MSC) equivalent to thrice the single dose was infused at week 6. Locomotor function, light and electron microscopy, immunohistochemistry and ex vivo diffusion tensor imaging were performed. Repeated infusion of MSCs (MSC-3) provided the greatest functional recovery compared to single and single high-dose infusions. The density of remyelinated axons in the injured spinal cord was the greatest in the MSC-3 group, followed by the MSC-1, HD-MSC and vehicle groups. Increased sprouting of the corticospinal tract and serotonergic axon density was the greatest in the MSC-3 group, followed by MSC-1, HD-MSC, and vehicle groups. Repeated infusion of MSCs over three weeks resulted in greater functional improvement than single administration of MSCs, even when the number of infused cells was tripled. MSC-treated rats showed axonal sprouting and remyelination in the chronic phase of SCI.
Collapse
Affiliation(s)
- Kota Kurihara
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan; Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Masanori Sasaki
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan; Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA.
| | - Hiroshi Nagahama
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan; Division of Radioisotope Research, Biomedical Research, Education and Instrumentation Center, Sapporo Medical University School of Medicine, Sapporo, Hokkaido 060-8556, Japan
| | - Hisashi Obara
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan; Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Ryunosuke Fukushi
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan; Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Ryosuke Hirota
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan; Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Mitsunori Yoshimoto
- Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Atsushi Teramoto
- Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Jeffery D Kocsis
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA; Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA; Center for Neuroscience and Regeneration Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Toshihiko Yamashita
- Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
| | - Osamu Honmou
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan; Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
4
|
Hirota R, Sasaki M, Kataoka-Sasaki Y, Oshigiri T, Kurihara K, Fukushi R, Oka S, Ukai R, Yoshimoto M, Kocsis JD, Yamashita T, Honmou O. Enhanced Network in Corticospinal Tracts after Infused Mesenchymal Stem Cells in Spinal Cord Injury. J Neurotrauma 2022; 39:1665-1677. [PMID: 35611987 PMCID: PMC9734021 DOI: 10.1089/neu.2022.0106] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Although limited spontaneous recovery occurs after spinal cord injury (SCI), current knowledge reveals that multiple forms of axon growth in spared axons can lead to circuit reorganization and a detour or relay pathways. This hypothesis has been derived mainly from studies of the corticospinal tract (CST), which is the primary descending motor pathway in mammals. The major CST is the dorsal CST (dCST), being the major projection from cortex to spinal cord. Two other components often called "minor" pathways are the ventral and the dorsal lateral CSTs, which may play an important role in spontaneous recovery. Intravenous infusion of mesenchymal stem cells (MSCs) provides functional improvement after SCI with an enhancement of axonal sprouting of CSTs. Detailed morphological changes of CST pathways, however, have not been fully elucidated. The primary objective was to evaluate detailed changes in descending CST projections in SCI after MSC infusion. The MSCs were infused intravenously one day after SCI. A combination of adeno-associated viral vector (AAV), which is an anterograde and non-transsynaptic axonal tracer, was injected 14 days after SCI induction. The AAV with advanced tissue clearing techniques were used to visualize the distribution pattern and high-resolution features of the individual axons coursing from above to below the lesion. The results demonstrated increased observable axonal connections between the dCST and axons in the lateral funiculus, both rostral and caudal to the lesion core, and an increase in observable axons in the dCST below the lesion. This increased axonal network could contribute to functional recovery by providing greater input to the spinal cord below the lesion.
Collapse
Affiliation(s)
- Ryosuke Hirota
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, and Sapporo Medical University School of Medicine, Sapporo, Japan.,Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Masanori Sasaki
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, and Sapporo Medical University School of Medicine, Sapporo, Japan.,Department of Neurology, and Yale University School of Medicine, New Haven, Connecticut, USA.,Address correspondence to: Masanori Sasaki, MD, PhD, Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, S1W17, Chuo-ku, Sapporo, Hokkaido 060-8556, Japan
| | - Yuko Kataoka-Sasaki
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, and Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Tsutomu Oshigiri
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, and Sapporo Medical University School of Medicine, Sapporo, Japan.,Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Kota Kurihara
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, and Sapporo Medical University School of Medicine, Sapporo, Japan.,Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Ryunosuke Fukushi
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, and Sapporo Medical University School of Medicine, Sapporo, Japan.,Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Shinichi Oka
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, and Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Ryo Ukai
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, and Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Mitsunori Yoshimoto
- Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Jeffery D. Kocsis
- Department of Neurology, and Yale University School of Medicine, New Haven, Connecticut, USA.,Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut, USA.,Center for Neuroscience and Regeneration Research, VA Connecticut Healthcare System, West Haven, Connecticut, USA
| | - Toshihiko Yamashita
- Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Osamu Honmou
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, and Sapporo Medical University School of Medicine, Sapporo, Japan.,Department of Neurology, and Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
5
|
Sheykhhasan M, Poondla N. Bone marrow mesenchymal stem cell treatment improves post-stroke cerebral function recovery by regulating gut microbiota in rats. World J Stem Cells 2022; 14:680-683. [PMID: 36157909 PMCID: PMC9453271 DOI: 10.4252/wjsc.v14.i8.680] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 06/29/2022] [Accepted: 08/18/2022] [Indexed: 02/07/2023] Open
Abstract
Early intervention with bone marrow mesenchymal stem cells to change the form and function of the gut microbiota may help rats regain neurological function after a stroke.
Collapse
Affiliation(s)
- Mohsen Sheykhhasan
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan 6517838695, Iran
| | - Naresh Poondla
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| |
Collapse
|
6
|
Takemura M, Sasaki M, Kataoka-Sasaki Y, Kiyose R, Nagahama H, Oka S, Ukai R, Yokoyama T, Kocsis JD, Ueba T, Honmou O. Repeated intravenous infusion of mesenchymal stem cells for enhanced functional recovery in a rat model of chronic cerebral ischemia. J Neurosurg 2022; 137:402-411. [PMID: 34861644 DOI: 10.3171/2021.8.jns21687] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 08/09/2021] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Stroke is a major cause of long-term disability, and there are few effective treatments that improve function in patients during the chronic phase of stroke. Previous research has shown that single systemic infusion of mesenchymal stem cells (MSCs) improves motor function in acute and chronic cerebral ischemia models in rats. A possible mechanism that could explain such an event includes the enhanced neural connections between cerebral hemispheres that contribute to therapeutic effects. In the present study, repeated infusions (3 times at weekly intervals) of MSCs were administered in a rat model of chronic stroke to determine if multiple dosing facilitated plasticity in neural connections. METHODS The authors induced middle cerebral artery occlusion (MCAO) in rats and, 8 weeks thereafter, used them as a chronic stroke model. The rats with MCAO were randomized and intravenously infused with vehicle only (vehicle group); with MSCs at week 8 (single administration: MSC-1 group); or with MSCs at weeks 8, 9, and 10 (3 times, repeated administration: MSC-3 group) via femoral veins. Ischemic lesion volume and behavioral performance were examined. Fifteen weeks after induction of MCAO, the thickness of the corpus callosum (CC) was determined using Nissl staining. Immunohistochemical analysis of the CC was performed using anti-neurofilament antibody. Interhemispheric connections through the CC were assessed ex vivo by diffusion tensor imaging. RESULTS Motor recovery was better in the MSC-3 group than in the MSC-1 group. In each group, there was no change in the ischemic volume before and after infusion. However, both thickness and optical density of neurofilament staining in the CC were greater in the MSC-3 group, followed by the MSC-1 group, and then the vehicle group. The increased thickness and optical density of neurofilament in the CC correlated with motor function at 15 weeks following induction of MCAO. Preserved neural tracts that ran through interhemispheric connections via the CC were also more extensive in the MSC-3 group, followed by the MSC-1 group and then the vehicle group, as observed ex vivo using diffusion tensor imaging. CONCLUSIONS These results indicate that repeated systemic administration of MSCs over 3 weeks resulted in greater functional improvement as compared to single administration and/or vehicle infusion. In addition, administration of MSCs is associated with promotion of interhemispheric connectivity through the CC in the chronic phase of cerebral infarction.
Collapse
Affiliation(s)
- Mitsuhiro Takemura
- 1Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine
- 3Department of Neurosurgery, Kochi Medical School, Kochi University, Nankoku, Kochi, Japan
| | - Masanori Sasaki
- 1Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine
- Departments of4Neurology and
| | - Yuko Kataoka-Sasaki
- 1Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine
| | - Ryo Kiyose
- 1Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine
| | - Hiroshi Nagahama
- 1Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine
- 2Division of Radioisotope Research, Biomedical Research, Education and Instrumentation Center, Sapporo Medical University School of Medicine, Sapporo, Hokkaido
| | - Shinichi Oka
- 1Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine
| | - Ryo Ukai
- 1Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine
| | - Takahiro Yokoyama
- 1Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine
| | - Jeffery D Kocsis
- Departments of4Neurology and
- 5Neuroscience, Yale University School of Medicine, New Haven; and
- 6Center for Neuroscience and Regeneration Research, VA Connecticut Healthcare System, West Haven, Connecticut
| | - Tetsuya Ueba
- 3Department of Neurosurgery, Kochi Medical School, Kochi University, Nankoku, Kochi, Japan
| | - Osamu Honmou
- 1Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine
- Departments of4Neurology and
| |
Collapse
|
7
|
Oka S, Yamaki T, Sasaki M, Ukai R, Takemura M, Yokoyama T, Kataoka-Sasaki Y, Onodera R, Ito YM, Kobayashi S, Kocsis JD, Iwadate Y, Honmou O. Intravenous infusion of auto serum-expanded autologous mesenchymal stem cells in chronic brain injury patients: a study protocol for a Phase II trial (Preprint). JMIR Res Protoc 2022; 11:e37898. [PMID: 35793128 PMCID: PMC9301565 DOI: 10.2196/37898] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/31/2022] [Accepted: 05/31/2022] [Indexed: 11/13/2022] Open
Abstract
Background Objective Methods Results Conclusions Trial Registration International Registered Report Identifier (IRRID)
Collapse
Affiliation(s)
- Shinichi Oka
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Advanced Regenerative Therapeutics, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Tomohiro Yamaki
- Division of Neurosurgery, Rehabilitation Center for Traumatic Apallics Chiba, National Agency for Automotive Safety and Victims' Aid, Chiba, Japan
| | - Masanori Sasaki
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Advanced Regenerative Therapeutics, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States
| | - Ryo Ukai
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Mitsuhiro Takemura
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takahiro Yokoyama
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yuko Kataoka-Sasaki
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Advanced Regenerative Therapeutics, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Rie Onodera
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yoichi M Ito
- Data Science Center, Institute of Health Science Innovation for Medical Care, Hokkaido University Hospital, Sapporo, Japan
| | - Shigeki Kobayashi
- Division of Neurosurgery, Rehabilitation Center for Traumatic Apallics Chiba, National Agency for Automotive Safety and Victims' Aid, Chiba, Japan
| | - Jeffery D Kocsis
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, United States
| | - Yasuo Iwadate
- Department of Neurological Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Osamu Honmou
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Advanced Regenerative Therapeutics, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
8
|
Brain Abnormalities in Pontine Infarction: A Longitudinal Diffusion Tensor Imaging and Functional Magnetic Resonance Imaging study. J Stroke Cerebrovasc Dis 2021; 31:106205. [PMID: 34879300 DOI: 10.1016/j.jstrokecerebrovasdis.2021.106205] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 10/15/2021] [Accepted: 10/21/2021] [Indexed: 01/19/2023] Open
Abstract
OBJECTIVES The aim of this study was to make a reasonable and accurate assessment of the prognosis of patients with pontine infarction. We assessed the changes in structure and function in the whole brain after pontine infarction from the acute to chronic phase using diffustion tensor imaging and functional magnetic resonance imaging. MATERIALS AND METHODS Sixteen individuals with a recent pontine infarction and sixteen healthy controls were recruited and underwent 3.0T DTI, resting-state fMRI and upper extremity Fugl-Myer (UE-FM) functional evaluation at five time points: within one week (T1), half a month (T2), one month (T3), three months (T4), and six months (T5) after onset. Tract-based spatial statistics was used to conduct a voxelwise analysis. RESULTS The fractional anisotropy (FA) values were significantly lower in the pontine infarction group than in the control group. Then, specific ROIs were analyzed. The FA values of 10 regions of interest were significantly increased at T2 compared with those at T1. The FA value of the corticospinal tract was significantly increased at T3 compared with that at T2. Regional brain activity results showed that the amplitude of low frequency fluctuations value of the frontal lobe decreased at T1, then increased. Finally, The UE-FM scores showed the same increased trend. CONCLUSION These findings show that the microstructure changes most significantly within half a month after pontine infarction and stabilizes after one month. The recovery of motor function in the later period is mainly caused by changes in the cortex. This facilitates more treatment options.
Collapse
|
9
|
Otsuka T, Maeda Y, Kurose T, Nakagawa K, Mitsuhara T, Kawahara Y, Yuge L. Comparisons of Neurotrophic Effects of Mesenchymal Stem Cells Derived from Different Tissues on Chronic Spinal Cord Injury Rats. Stem Cells Dev 2021; 30:865-875. [PMID: 34148410 DOI: 10.1089/scd.2021.0070] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Cell-based therapies with mesenchymal stem cells (MSCs) are considered as promising strategies for spinal cord injury (SCI). MSCs have unique characteristics due to differences in the derived tissues. However, relatively few studies have focused on differences in the therapeutic effects of MSCs derived from different tissues. In this study, the therapeutic effects of adipose tissue-derived MSCs, bone marrow-derived MSCs, and cranial bone-derived MSCs (cMSCs) on chronic SCI model rats were compared. MSCs were established from the collected adipose tissue, bone marrow, and cranial bone. Neurotrophic factor expression of each MSC type was analyzed by real-time PCR. SCI rats were established using the weight-drop method and transplanted intravenously with MSCs at 4 weeks after SCI. Hindlimb motor function was evaluated from before injury to 4 weeks after transplantation. Endogenous neurotrophic factor and neural repair factor expression in spinal cord (SC) tissue were examined by real-time PCR and western blot analyses. Although there were no differences in the expression levels of cell surface markers and multipotency, expression of Bdnf, Ngf, and Sort1 (Nt-3) was relatively higher in cMSCs. Transplantation of cMSCs improved motor function of chronic SCI model rats. Although there was no difference in the degree of engraftment of transplanted cells in the injured SC tissue, transplantation of cMSCs enhanced Bdnf, TrkB, and Gap-43 messenger RNA expression and synaptophysin protein expression in injured SC tissue. As compared with MSCs derived other tissues, cMSCs highly express many neurotrophic factors, which improved motor function in chronic SCI model rats by promoting endogenous neurotrophic and neural plasticity factors. These results demonstrate the efficacy of cMSCs in cell-based therapy for chronic SCI.
Collapse
Affiliation(s)
- Takashi Otsuka
- Division of Bio-Environmental Adaptation Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yuyo Maeda
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Tomoyuki Kurose
- Division of Bio-Environmental Adaptation Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kei Nakagawa
- Division of Bio-Environmental Adaptation Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Takafumi Mitsuhara
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | | | - Louis Yuge
- Division of Bio-Environmental Adaptation Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.,Space Bio-Laboratories Co., Ltd., Hiroshima, Japan
| |
Collapse
|
10
|
Atia MM, Alghriany AA. Adipose-derived mesenchymal stem cells rescue rat hippocampal cells from aluminum oxide nanoparticle-induced apoptosis via regulation of P53, Aβ, SOX2, OCT4, and CYP2E1. Toxicol Rep 2021; 8:1156-1168. [PMID: 34150525 PMCID: PMC8190131 DOI: 10.1016/j.toxrep.2021.06.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/01/2021] [Accepted: 06/02/2021] [Indexed: 12/20/2022] Open
Abstract
Mesenchymal stem cells (MSCs) possess a preventive capacity against free radical toxicity in various tissues. The present study aimed to demonstrate the reformative and treatment roles of adipose-derived MSCs (AD-MSCs) against severe toxicity in the hippocampal cells of the brain caused by aluminum oxide nanoparticles (Al2O3-NPs). Rats were divided into five experimental groups: an untreated control group, a control group receiving NaCl, a group receiving Al2O3-NPs (6 mg/kg) for 20 days, a group that was allowed to recover (R) for 20 days following treatment with Al2O3-NPs, and a Al2O3-NPs + AD-MSCs group, where each rat was injected with 0.8 × 106 AD-MSCs via the caudal vein. Oral administration of Al2O3-NPs increased the protein levels of P53, cleaved caspase-3, CYP2E1, and beta-amyloid (Aβ); contrarily, AD-MSCs transplantation downregulated the levels of these proteins. In addition, the AD-MSCs-treated hippocampal cells were protected from Al2O3-NPs-induced toxicity, as detected by the expression levels of Sox2 and Oct4 that are essential for the maintenance of self-renewal. It was also found that AD-MSCs injection significantly altered the levels of brain total peroxide and monoamine oxidase (MAO)-A and MAO-B activities. Histologically, our results indicated that AD-MSCs alleviated the severe damage in the hippocampal cells induced by Al2O3-NPs. Moreover, the role of AD-MSCs in reducing hippocampal cell death was reinforced by the regulation of P53, cleaved caspase-3, Aβ, and CYP2E1 proteins, as well as by the regulation of SOX2 and OCT4 levels and MAO-A and MAO-B activities.
Collapse
Key Words
- AD-MSCs, adipose-derived mesenchymal stem cells
- Adipose-Derived mesenchymal stem cells
- Al2O3-NPs, Aluminum oxide nanoparticles
- Aluminum oxide nanoparticles
- Apoptosis
- Aβ, amyloid beta
- EGTA, ethylene glycol tetraacetic acid
- Hippocampal cells
- MAO-A and B, monoamine oxidase A, B
- Oct4, octamer-binding transcription factor 4
- ROS, reactive oxygen species
- Sox2, sex-determining region Y-box 2
- TEM, transmission electron microscopy
Collapse
Affiliation(s)
- Mona M. Atia
- Laboratory of Molecular Cell Biology, Department of Zoology, Faculty of Science, Assiut University, Egypt
| | - Alshaimaa A.I. Alghriany
- Laboratory of Molecular Cell Biology, Department of Zoology, Faculty of Science, Assiut University, Egypt
| |
Collapse
|
11
|
Magota H, Sasaki M, Kataoka-Sasaki Y, Oka S, Ukai R, Kiyose R, Onodera R, Kocsis JD, Honmou O. Intravenous infusion of mesenchymal stem cells delays disease progression in the SOD1G93A transgenic amyotrophic lateral sclerosis rat model. Brain Res 2021; 1757:147296. [PMID: 33516815 DOI: 10.1016/j.brainres.2021.147296] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 12/20/2020] [Accepted: 01/10/2021] [Indexed: 12/14/2022]
Abstract
ALS is a devastating neurodegenerative disease with few curative strategies. Both sporadic and familial ALS display common clinical features that show progressive paralysis. The pathogenesis remains unclear, but disruption of the blood-spinal cord barrier (BSCB) may contribute to the degeneration of motor neurons. Thus, restoration of the disrupted BSCB and neuroprotection for degenerating motor neurons could be therapeutic targets. We tested the hypothesis that an intravenous infusion of MSCs would delay disease progression through the preservation of BSCB function and increased expression of a neurotrophic factor, neurturin, in SOD1G93A ALS rats. When the open-field locomotor function was under 16 on the Basso, Beattie, and Bresnahan (BBB) scoring scale, the rats were randomized into two groups; one received an intravenous infusion of MSCs, while the other received vehicle alone. Locomotor function was recorded using BBB scoring and rotarod testing. Histological analyses, quantitative reverse transcription-polymerase chain reaction (qRT-PCR), were performed. The MSC group exhibited reduced deterioration of locomotor activity compared to the vehicle group, which displayed progressive deterioration of hind limb function. We observed the protection of motor neuron loss and preservation of microvasculature using Evans blue leakage and immunohistochemical analyses in the MSC group. Confocal microscopy revealed infused green fluorescent protein+ (GFP+) MSCs in the spinal cord, and the GFP gene was detected by nested PCR. Neurturin expression levels were significantly higher in the MSC group. Thus, restoration of the BSCB and the protection of motor neurons might be contributing mechanisms to delay disease progression in SOD1G93A ALS rats.
Collapse
Affiliation(s)
- Hirotoshi Magota
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Hokkaido 060-8556, Japan; Tominaga Hospital, Naniwa-ku, Osaka-shi, Osaka 556-0017, Japan
| | - Masanori Sasaki
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Hokkaido 060-8556, Japan; Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, United States; Center for Neuroscience and Regeneration Research, VA Connecticut Healthcare System, West Haven, CT 06516, United States.
| | - Yuko Kataoka-Sasaki
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Hokkaido 060-8556, Japan
| | - Shinichi Oka
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Hokkaido 060-8556, Japan
| | - Ryo Ukai
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Hokkaido 060-8556, Japan
| | - Ryo Kiyose
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Hokkaido 060-8556, Japan; Tominaga Hospital, Naniwa-ku, Osaka-shi, Osaka 556-0017, Japan
| | - Rie Onodera
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Hokkaido 060-8556, Japan
| | - Jeffery D Kocsis
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, United States; Center for Neuroscience and Regeneration Research, VA Connecticut Healthcare System, West Haven, CT 06516, United States
| | - Osamu Honmou
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Hokkaido 060-8556, Japan; Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, United States; Center for Neuroscience and Regeneration Research, VA Connecticut Healthcare System, West Haven, CT 06516, United States
| |
Collapse
|
12
|
Intravenous Infusion of Mesenchymal Stem Cells Enhances Therapeutic Efficacy of Reperfusion Therapy in Cerebral Ischemia. World Neurosurg 2021; 149:e160-e169. [PMID: 33618048 DOI: 10.1016/j.wneu.2021.02.056] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/10/2021] [Accepted: 02/11/2021] [Indexed: 01/02/2023]
Abstract
OBJECTIVE Reperfusion therapy is a standard therapeutic strategy for acute stroke. Non-favorable outcomes are thought to partially result from impaired microcirculatory flow in ischemic tissue. Intravenous infusion of mesenchymal stem cells (MSCs) reduces stroke volume and improves behavioral function in stroke. One suggested therapeutic mechanism is the restoration of the microvasculature. The goal of this study was to determine whether infused MSCs enhance the therapeutic efficacy of reperfusion therapy following stroke in rats. METHODS First, to establish a transient middle cerebral artery occlusion (MCAO) model displaying approximately identical neurologic function and lesion volume as seen in permanent MCAO (pMCAO) at day 7 after stroke induction, we transiently occluded the MCA for 90, 110, and 120 minutes. We found that the 110-minute occlusion met these criteria and was used as the transient MCAO (tMCAO) model. Next, 4 MCAO groups were used to compare the therapeutic efficacy of infused MSCs: (1) pMCAO+vehicle, (2) tMCAO+vehicle, (3) pMCAO+MSC, and (4) tMCAO+MSC. Our ischemic model was a unique ischemic model system in which both pMCAO and tMCAO provided similar outcomes during the study period in the groups without MSC infusion groups. Behavioral performance, ischemic volume, and regional cerebral blood flow (rCBF) using arterial spin labeling-magnetic resonance imaging and histologic evaluation of microvasculature was performed. RESULTS The behavioral function, rCBF, and restoration of microvasculature were greater in group 4 than in group 3. Thus, infused MSCs facilitated the therapeutic efficacy of MCA reperfusion in this rat model system. CONCLUSIONS Intravenous infusion of MSCs may enhance therapeutic efficacy of reperfusion therapy.
Collapse
|
13
|
Honmou O, Yamashita T, Morita T, Oshigiri T, Hirota R, Iyama S, Kato J, Sasaki Y, Ishiai S, Ito YM, Namioka A, Namioka T, Nakazaki M, Kataoka-Sasaki Y, Onodera R, Oka S, Sasaki M, Waxman SG, Kocsis JD. Intravenous infusion of auto serum-expanded autologous mesenchymal stem cells in spinal cord injury patients: 13 case series. Clin Neurol Neurosurg 2021; 203:106565. [PMID: 33667953 DOI: 10.1016/j.clineuro.2021.106565] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 11/12/2020] [Accepted: 02/16/2021] [Indexed: 01/24/2023]
Abstract
BACKGROUND Although spinal cord injury (SCI) is a major cause of disability, current therapeutic options remain limited. Recent progress in cellular therapy with mesenchymal stem cells (MSCs) has provided improved function in animal models of SCI. We investigated the safety and feasibility of intravenous infusion of MSCs for SCI patients and assessed functional status after MSC infusion. METHODS In this phase 2 study of intravenous infusion of autologous MSCs cultured in auto-serum, a single infusion of MSCs under Good Manufacturing Practice (GMP) production was delivered in 13 SCI patients. In addition to assessing feasibility and safety, neurological function was assessed using the American Spinal Injury Association Impairment Scale (ASIA), International Standards for Neurological and Functional Classification of Spinal Cord (ISCSCI-92). Ability of daily living was assessed using Spinal Cord Independence Measure (SCIM-III). The study protocol was based on advice provided by the Pharmaceuticals and Medical Devices Agency in Japan. The trial was registered with the Japan Medical Association (JMA-IIA00154). RESULTS No serious adverse events were associated with MSC injection. There was neurologic improvement based on ASIA grade in 12 of the 13 patients at six months post-MSC infusion. Five of six patients classified as ASIA A prior to MSC infusion improved to ASIA B (3/6) or ASIA C (2/6), two ASIA B patients improved to ASIA C (1/2) or ASIA D (1/2), five ASIA C patients improved and reached a functional status of ASIA D (5/5). Notably, improvement from ASIA C to ASIA D was observed one day following MSC infusion for all five patients. Assessment of both ISCSCI-92, SCIM-III also demonstrated functional improvements at six months after MSC infusion, compared to the scores prior to MSC infusion in all patients. CONCLUSION While we emphasize that this study was unblinded, and does not exclude placebo effects or a contribution of endogenous recovery or observer bias, our observations provide evidence supporting the feasibility, safety and functional improvements of infused MSCs into patients with SCI.
Collapse
Affiliation(s)
- Osamu Honmou
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Hokkaido, Japan; Department of Advanced Regenerative Therapeutics, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Hokkaido, Japan; Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Toshihiko Yamashita
- Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Hokkaido, Japan
| | - Tomonori Morita
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Hokkaido, Japan; Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Hokkaido, Japan
| | - Tsutomu Oshigiri
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Hokkaido, Japan; Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Hokkaido, Japan
| | - Ryosuke Hirota
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Hokkaido, Japan; Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Hokkaido, Japan
| | - Satoshi Iyama
- Department of Hematology, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Hokkaido, Japan
| | - Junji Kato
- Department of Medical Oncology, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Hokkaido, Japan
| | - Yuichi Sasaki
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Hokkaido, Japan; Department of Rehabilitation, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Hokkaido, Japan
| | - Sumio Ishiai
- Department of Rehabilitation, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Hokkaido, Japan
| | - Yoichi M Ito
- Biostatistics Division, Hokkaido University Hospital Clinical Research and Medical Innovation Center, Sapporo, Hokkaido, 060-8648, Japan
| | - Ai Namioka
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Hokkaido, Japan
| | - Takahiro Namioka
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Hokkaido, Japan
| | - Masahito Nakazaki
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Hokkaido, Japan
| | - Yuko Kataoka-Sasaki
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Hokkaido, Japan; Department of Advanced Regenerative Therapeutics, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Hokkaido, Japan
| | - Rie Onodera
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Hokkaido, Japan
| | - Shinichi Oka
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Hokkaido, Japan; Department of Advanced Regenerative Therapeutics, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Hokkaido, Japan
| | - Masanori Sasaki
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Hokkaido, Japan; Department of Advanced Regenerative Therapeutics, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Hokkaido, Japan; Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA.
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Jeffery D Kocsis
- Department of Neurology, Yale University School of Medicine, New Haven, CT, 06510, USA
| |
Collapse
|
14
|
Nagahama H, Sasaki M, Kiyose R, Yasuda N, Honmou O. [3. Magnetic Resonance Imaging for Analysis of Neural Plasticity Induced by Neuroregenerative Therapy]. Nihon Hoshasen Gijutsu Gakkai Zasshi 2021; 77:1238-1244. [PMID: 34670933 DOI: 10.6009/jjrt.2021_jsrt_77.10.1238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Affiliation(s)
- Hiroshi Nagahama
- Division of Radioisotope Research, Biomedical Research, Education and Instrumentation Center, Sapporo Medical University School of Medicine
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine
| | - Masanori Sasaki
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine
| | - Ryo Kiyose
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine
| | - Naomi Yasuda
- Department of Cardiovascular Surgery, Sapporo Medical University School of Medicine
| | - Osamu Honmou
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine
| |
Collapse
|
15
|
Intravenous delivery of mesenchymal stem cells protects both white and gray matter in spinal cord ischemia. Brain Res 2020; 1747:147040. [DOI: 10.1016/j.brainres.2020.147040] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 07/10/2020] [Accepted: 07/27/2020] [Indexed: 12/11/2022]
|
16
|
Angeloni C, Gatti M, Prata C, Hrelia S, Maraldi T. Role of Mesenchymal Stem Cells in Counteracting Oxidative Stress-Related Neurodegeneration. Int J Mol Sci 2020; 21:ijms21093299. [PMID: 32392722 PMCID: PMC7246730 DOI: 10.3390/ijms21093299] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 04/30/2020] [Accepted: 05/04/2020] [Indexed: 02/07/2023] Open
Abstract
Neurodegenerative diseases include a variety of pathologies such as Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, amyotrophic lateral sclerosis, and so forth, which share many common characteristics such as oxidative stress, glycation, abnormal protein deposition, inflammation, and progressive neuronal loss. The last century has witnessed significant research to identify mechanisms and risk factors contributing to the complex etiopathogenesis of neurodegenerative diseases, such as genetic, vascular/metabolic, and lifestyle-related factors, which often co-occur and interact with each other. Apart from several environmental or genetic factors, in recent years, much evidence hints that impairment in redox homeostasis is a common mechanism in different neurological diseases. However, from a pharmacological perspective, oxidative stress is a difficult target, and antioxidants, the only strategy used so far, have been ineffective or even provoked side effects. In this review, we report an analysis of the recent literature on the role of oxidative stress in Alzheimer’s and Parkinson’s diseases as well as in amyotrophic lateral sclerosis, retinal ganglion cells, and ataxia. Moreover, the contribution of stem cells has been widely explored, looking at their potential in neuronal differentiation and reporting findings on their application in fighting oxidative stress in different neurodegenerative diseases. In particular, the exposure to mesenchymal stem cells or their secretome can be considered as a promising therapeutic strategy to enhance antioxidant capacity and neurotrophin expression while inhibiting pro-inflammatory cytokine secretion, which are common aspects of neurodegenerative pathologies. Further studies are needed to identify a tailored approach for each neurodegenerative disease in order to design more effective stem cell therapeutic strategies to prevent a broad range of neurodegenerative disorders.
Collapse
Affiliation(s)
- Cristina Angeloni
- School of Pharmacy, University of Camerino, Via Gentile III da Varano, 62032 Camerino, Italy;
| | - Martina Gatti
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Via del Pozzo 71, 41124 Modena, Italy; (M.G.); (T.M.)
| | - Cecilia Prata
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum—University of Bologna, Via Irnerio 48, 40126 Bologna, Italy
- Correspondence:
| | - Silvana Hrelia
- Department for Life Quality Studies, Alma Mater Studiorum—University of Bologna, Corso d’Augusto 237, 47921 Rimini, Italy;
| | - Tullia Maraldi
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Via del Pozzo 71, 41124 Modena, Italy; (M.G.); (T.M.)
| |
Collapse
|
17
|
“Chronic” State in Neural Diseases as the Target of Cellular Therapy with Mesenchymal Stem Cells. World Neurosurg 2020; 135:375-376. [DOI: 10.1016/j.wneu.2019.12.137] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
18
|
Surugiu R, Olaru A, Hermann DM, Glavan D, Catalin B, Popa-Wagner A. Recent Advances in Mono- and Combined Stem Cell Therapies of Stroke in Animal Models and Humans. Int J Mol Sci 2019; 20:ijms20236029. [PMID: 31795466 PMCID: PMC6928803 DOI: 10.3390/ijms20236029] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 11/24/2019] [Accepted: 11/29/2019] [Indexed: 12/14/2022] Open
Abstract
Following the failure of acute neuroprotection therapies, major efforts are currently made worldwide to promote neurological recovery and brain plasticity in the subacute and post-acute phases of stroke. Currently, there is hope that stroke recovery might be promoted by cell-based therapies. The field of stem cell therapy for cerebral ischemia has made significant progress in the last five years. A variety of stem cells have been tested in animal models and humans including adipose stem cells, human umbilical cord blood-derived mesenchymal stem cells, human amnion epithelial cells, human placenta amniotic membrane-derived mesenchymal stem cells, adult human pluripotent-like olfactory stem cells, human bone marrow endothelial progenitor cells, electrically-stimulated human neuronal progenitor cells, or induced pluripotent stem cells (iPSCs) of human origin. Combination therapies in animal models include a mix of two or more therapeutic factors consisting of bone marrow stromal cells, exercise and thyroid hormones, endothelial progenitor cells overexpressing the chemokine CXCL12. Mechanisms underlying the beneficial effects of transplanted cells include the “bystander” effects, paracrine mechanisms, or extracellular vesicles-mediated restorative effects. Mitochondria transfer also appears to be a powerful strategy for regenerative processes. Studies in humans are currently limited to a small number of studies using autologous stem cells mainly aimed to assess tolerability and side-effects of human stem cells in the clinic.
Collapse
Affiliation(s)
- Roxana Surugiu
- Center of Clinical and Experimental Medicine, University of Medicine and Pharmacy, 20049 Craiova, Romania
| | - Andrei Olaru
- Department of Ophthalmology, University of Medicine and Pharmacy, 20049 Craiova, Romania
| | - Dirk M Hermann
- Chair of Vascular Neurology, Dementia and Ageing Research, Department of Neurology, University of Duisburg-Essen, University Hospital Essen, 45122 Essen, Germany
| | - Daniela Glavan
- Department of Psychiatry, University of Medicine and Pharmacy, 20049 Craiova, Romania
| | - Bogdan Catalin
- Experimental Research Centre for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, 20049 Craiova, Romania
| | - Aurel Popa-Wagner
- Chair of Vascular Neurology, Dementia and Ageing Research, Department of Neurology, University of Duisburg-Essen, University Hospital Essen, 45122 Essen, Germany
- Griffith University Menzies Health Institute of Queensland, Gold Coast Campus and Queensland Eye Institute, Brisbane, QLD 4000, Australia
| |
Collapse
|
19
|
Crofts A, Kelly ME, Gibson CL. Imaging Functional Recovery Following Ischemic Stroke: Clinical and Preclinical fMRI Studies. J Neuroimaging 2019; 30:5-14. [PMID: 31608550 PMCID: PMC7003729 DOI: 10.1111/jon.12668] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 09/12/2019] [Accepted: 09/25/2019] [Indexed: 12/18/2022] Open
Abstract
Disability and effectiveness of physical therapy are highly variable following ischemic stroke due to different brain regions being affected. Functional magnetic resonance imaging (fMRI) studies of patients in the months and years following stroke have given some insight into how the brain recovers lost functions. Initially, new pathways are recruited to compensate for the lost region, showing as a brighter blood oxygen‐level‐dependent (BOLD) signal over a larger area during a task than in healthy controls. Subsequently, activity is reduced to baseline levels as pathways become more efficient, mimicking the process of learning typically seen during development. Preclinical models of ischemic stroke aim to enhance understanding of the biology underlying recovery following stroke. However, the pattern of recruitment and focusing seen in humans has not been observed in preclinical fMRI studies that are highly variable methodologically. Resting‐state fMRI studies show more consistency; however, there are still confounding factors to address. Anesthesia and method of stroke induction are the two main sources of variability in preclinical studies; improvements here can reduce variability and increase the intensity and reproducibility of the BOLD response detected by fMRI. Differences in task or stimulus and differences in analysis method also present a source of variability. This review compares clinical and preclinical fMRI studies of recovery following stroke and focuses on how refinement of preclinical models and MRI methods may obtain more representative fMRI data in relation to human studies.
Collapse
Affiliation(s)
- Andrew Crofts
- Department of Neuroscience, Psychology and Behaviour, University of Leicester, Leicester, UK
| | - Michael E Kelly
- Preclinical Imaging Facility, Core Biotechnology Services, University of Leicester, Leicester, UK
| | - Claire L Gibson
- School of Psychology, University of Nottingham, Nottingham, UK
| |
Collapse
|
20
|
Nakazaki M, Oka S, Sasaki M, Kataoka-Sasaki Y, Onodera R, Komatsu K, Iihoshi S, Hiroura M, Kawaguchi A, Kocsis JD, Honmou O. Prevention of neointimal hyperplasia induced by an endovascular stent via intravenous infusion of mesenchymal stem cells. J Neurosurg 2019; 133:1773-1785. [PMID: 31585431 DOI: 10.3171/2019.7.jns19575] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 07/01/2019] [Indexed: 11/06/2022]
Abstract
OBJECTIVE In-stent restenosis after percutaneous transluminal angioplasty and stenting (PTAS) due to neointimal hyperplasia is a potential cause of clinical complications, including repeated revascularization and ischemic events. Neointimal hyperplasia induced by an inflammatory response to the stent strut may be a possible mechanism of in-stent restenosis. Intravenous infusion of bone marrow-derived mesenchymal stem cells (MSCs) has been reported to show therapeutic efficacy for cerebral stroke, presumably by an antiinflammatory effect. This study aimed to determine whether MSCs can reduce or prevent neointimal hyperplasia induced by an endovascular stent. METHODS In this study, two types of bare metal stents were deployed using a porcine (mini-pig) model. One stent was implanted in the common carotid artery (CCA), which is considered quite similar to the human CCA, and the other was inserted in the superficial cervical artery (SCA), which is similar in size to the human middle cerebral artery. Angiographic images, intravascular ultrasound (IVUS) imaging, and microscopic images were used for analysis. RESULTS Angiographic images and IVUS studies revealed that intravenous infusion of MSCs immediately after deployment of stents prevented in-stent stenosis of the CCA and SCA. Histological analysis also confirmed that inflammatory responses around the stent struts were reduced in both the stented CCA and SCA in the mini-pig. CONCLUSIONS Intravenous infusion of MSCs inhibited the inflammatory reaction to an implanted stent strut, and prevented progressive neointimal hyperplasia in the stented CCA and SCA in a porcine model. Thus, MSC treatment could attenuate the recurrence of cerebral ischemic events after stenting.
Collapse
Affiliation(s)
- Masahito Nakazaki
- 1Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, and
- 4Department of Neurology, Yale University School of Medicine, New Haven, Connecticut; and
- 5Center for Neuroscience and Regeneration Research, VA Connecticut Healthcare System, West Haven, Connecticut
| | - Shinichi Oka
- 1Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, and
| | - Masanori Sasaki
- 1Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, and
- 4Department of Neurology, Yale University School of Medicine, New Haven, Connecticut; and
- 5Center for Neuroscience and Regeneration Research, VA Connecticut Healthcare System, West Haven, Connecticut
| | - Yuko Kataoka-Sasaki
- 1Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, and
| | - Rie Onodera
- 1Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, and
| | - Katsuya Komatsu
- 2Department of Neurosurgery, Sapporo Medical University School of Medicine, Sapporo, Hokkaido
| | - Satoshi Iihoshi
- 2Department of Neurosurgery, Sapporo Medical University School of Medicine, Sapporo, Hokkaido
| | - Manabu Hiroura
- 3NIPRO Life Science Site, NIPRO Corporation, Kusatsu, Shiga, Japan
| | - Akira Kawaguchi
- 3NIPRO Life Science Site, NIPRO Corporation, Kusatsu, Shiga, Japan
| | - Jeffery D Kocsis
- 4Department of Neurology, Yale University School of Medicine, New Haven, Connecticut; and
- 5Center for Neuroscience and Regeneration Research, VA Connecticut Healthcare System, West Haven, Connecticut
| | - Osamu Honmou
- 1Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, and
- 4Department of Neurology, Yale University School of Medicine, New Haven, Connecticut; and
- 5Center for Neuroscience and Regeneration Research, VA Connecticut Healthcare System, West Haven, Connecticut
| |
Collapse
|
21
|
Jin J, Tang Y, Li K, Zuo X, Zhan L, Sun W, Xu E. Bone Marrow Stromal Cells Alleviate Secondary Damage in the Substantia Nigra After Focal Cerebral Infarction in Rats. Front Cell Neurosci 2019; 13:338. [PMID: 31396057 PMCID: PMC6668054 DOI: 10.3389/fncel.2019.00338] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 07/10/2019] [Indexed: 12/02/2022] Open
Abstract
Transplantation of bone marrow stromal cells (BMSCs) is a promising therapy for ischemic stroke. Previously, we had reported that the secondary degeneration occurred in the ipsilateral substantia nigra (SN) after permanent distal branch of middle cerebral artery occlusion (dMCAO) in Sprague-Dawley rats. However, whether BMSCs have neurorestorative effects on the secondary damage in the SN after focal cerebral infarction has not known. In this study, rats were subjected to dMCAO followed by intravenous administration of BMSCs 1 day later. We found that transplanted BMSCs survived and migrated to cortical infarct areas and ipsilateral SN. Furthermore, BMSCs promoted neurogenesis through proliferation and differentiation in the SN after dMCAO. Rats implanted with BMSCs showed significant improvement in their performance of modified neurological severity scores and adhesive-removal test. Engrafted BMSCs enhanced survival of dopaminergic neuron, reduced gliosis in the ipsilateral SN, and increased contents of dopamine (DA) and its metabolites in the ipsilateral striatum after dMCAO. With pseudorabies virus-152 as a retrograde tracer, we also demonstrated that BMSCs could effectively enhance the cortico-striatum-nigral connections. These results suggest that BMSCs transplantation exerts neurorestorative effects after cortical infarction through promoting endogenous neurogenesis, increasing contents of DA and its metabolites, alleviating the secondary neuronal damage in the SN, enhancing the cortico-striatum-nigral projections pathway, and finally improving the neurological functional outcome.
Collapse
Affiliation(s)
- Jizi Jin
- Department of Neurology, Institute of Neurosciences, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province, Ministry of Education of China, Collaborative Innovation Center for Neurogenetics and Channelopathies, Guangzhou, China
| | - Yanyan Tang
- Department of Neurology, Institute of Neurosciences, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province, Ministry of Education of China, Collaborative Innovation Center for Neurogenetics and Channelopathies, Guangzhou, China
| | - Kongping Li
- Department of Neurology, Institute of Neurosciences, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province, Ministry of Education of China, Collaborative Innovation Center for Neurogenetics and Channelopathies, Guangzhou, China
| | - Xialin Zuo
- Department of Neurology, Institute of Neurosciences, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province, Ministry of Education of China, Collaborative Innovation Center for Neurogenetics and Channelopathies, Guangzhou, China
| | - Lixuan Zhan
- Department of Neurology, Institute of Neurosciences, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province, Ministry of Education of China, Collaborative Innovation Center for Neurogenetics and Channelopathies, Guangzhou, China
| | - Weiwen Sun
- Department of Neurology, Institute of Neurosciences, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province, Ministry of Education of China, Collaborative Innovation Center for Neurogenetics and Channelopathies, Guangzhou, China
| | - En Xu
- Department of Neurology, Institute of Neurosciences, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province, Ministry of Education of China, Collaborative Innovation Center for Neurogenetics and Channelopathies, Guangzhou, China
| |
Collapse
|
22
|
Stem Cells to Modulate IR: a Regenerative Medicine-Based Approach to Organ Preservation. CURRENT TRANSPLANTATION REPORTS 2019. [DOI: 10.1007/s40472-019-00240-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|