1
|
Mikuni M, Horiuchi K, Ishikura A, Kimura S, Masutani S, Watanabe S, Mikami A, Ishikawa S, Narita H, Kusumi I, Sasaki H. Suspected Postpartum Depression Revealed to be CSF1R-Related Leukoencephalopathy: A Case Report. Case Rep Neurol 2024; 16:281-287. [PMID: 39483843 PMCID: PMC11527462 DOI: 10.1159/000541551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 09/18/2024] [Indexed: 11/03/2024] Open
Abstract
Introduction This is a case of a 32-year-old woman who developed postpartum depression (PPD). She became anxious and depressive about caring for her child, and the Edinburgh Postnatal Depression Scale (EPDS) test showed a score of 9 at 2 weeks after delivery, and at 7 months postpartum, she presented with major melancholic depression followed by mild cognitive decline without any neurological symptoms except cluttering speech. Case Presentation Cerebral magnetic resonance imaging showed confluent fluid-attenuated inversion recovery hyperintensities in the periventricular and frontal deep white matter, with multiple spotty calcifications in the frontal white matter by cerebral CT. Genetic testing revealed a mutation in the colony-stimulating factor 1 receptor (CSF1R). Conclusion This case report is consistent with evidence that PPD may have organic causes in some cases, including CSF1R mutations. Atypical findings such as mild cognitive decline combined with PPD in psychiatric interview may justify brain imaging to avoid misdiagnosis, since CSF1R-related leukoencephalopathy is probably an under-recognized disease in medical psychiatry. Further investigations are needed to clarify a pathophysiological correlation between CSF1R signaling abnormality and PPD as well as major depression.
Collapse
Affiliation(s)
- Masahiko Mikuni
- Department of Psychiatry, Hakodate Watanabe Hospital, Hakodate, Japan
- Department of Psychiatry, Hokkaido University Graduate School of Medicine, Sapporo, Japan
- Department of Psychiatry and Human Behavior, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Kazuhiro Horiuchi
- Department of Neurology, Hakodate Municipal Hospital, Hakodate, Japan
| | - Ayako Ishikura
- Department of Pediatrics, Hakodate Central General Hospital, Hakodate, Japan
| | - Soichiro Kimura
- Department of Psychiatry, Hakodate Watanabe Hospital, Hakodate, Japan
| | - Sho Masutani
- Department of Psychiatry, Hakodate Watanabe Hospital, Hakodate, Japan
| | - Shinya Watanabe
- Department of Psychiatry, Hakodate Watanabe Hospital, Hakodate, Japan
| | - Akihiro Mikami
- Department of Psychiatry, Hakodate Watanabe Hospital, Hakodate, Japan
| | - Shuhei Ishikawa
- Department of Psychiatry, Hokkaido University Hospital, Sapporo, Japan
| | - Hisashi Narita
- Department of Psychiatry, Hokkaido University Hospital, Sapporo, Japan
| | - Ichiro Kusumi
- Department of Psychiatry, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Hidenao Sasaki
- Department of Neurology, Hakodate Central General Hospital, Hakodate, Japan
| |
Collapse
|
2
|
Lee HY, Moon SH, Kang D, Choi E, Yang GH, Kim KN, Won JY, Yi S. A multi-channel collagen conduit with aligned Schwann cells and endothelial cells for enhanced neuronal regeneration in spinal cord injury. Biomater Sci 2023; 11:7884-7896. [PMID: 37906468 DOI: 10.1039/d3bm01152f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Traumatic spinal cord injury (SCI) leads to Wallerian degeneration and the accompanying disruption of vasculature leads to ischemia, which damages motor and sensory function. Therefore, understanding the biological environment during regeneration is essential to promote neuronal regeneration and overcome this phenomenon. The band of Büngner is a structure of an aligned Schwann cell (SC) band that guides axon elongation providing a natural recovery environment. During axon elongation, SCs promote axon elongation while migrating along neovessels (endothelial cells [ECs]). To model this, we used extrusion 3D bioprinting to develop a multi-channel conduit (MCC) using collagen for the matrix region and sacrificial alginate to make the channel. The MCC was fabricated with a structure in which SCs and ECs were longitudinally aligned to mimic the sophisticated recovering SCI conditions. Also, we produced an MCC with different numbers of channels. The aligned SCs and ECs in the 9-channel conduit (9MCC-SE) were more biocompatible and led to more proliferation than the 5-channel conduit (5MCC-SE) in vitro. Also, the 9MCC-SE resulted in a greater healing effect than the 5MCC-SE with respect to neuronal regeneration, remyelination, inflammation, and angiogenesis in vivo. The above tissue recovery results led to motor function repair. Our results show that our 9MCC-SE model represents a new therapeutic strategy for SCI.
Collapse
Affiliation(s)
- Hye Yeong Lee
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, 134 Sinchon-dong, Seodaemun-gu, Seoul 03722, Republic of Korea.
| | - Seo Hyun Moon
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, 134 Sinchon-dong, Seodaemun-gu, Seoul 03722, Republic of Korea.
| | - Donggu Kang
- Research Institute of Additive Manufacturing and Regenerative Medicine, Baobab Healthcare Inc., 55 Hanyangdaehak-Ro, Ansan, Gyeonggi-Do, 15588, South Korea
| | - Eunjeong Choi
- Research Institute of Additive Manufacturing and Regenerative Medicine, Baobab Healthcare Inc., 55 Hanyangdaehak-Ro, Ansan, Gyeonggi-Do, 15588, South Korea
| | - Gi Hoon Yang
- Research Institute of Additive Manufacturing and Regenerative Medicine, Baobab Healthcare Inc., 55 Hanyangdaehak-Ro, Ansan, Gyeonggi-Do, 15588, South Korea
| | - Keung Nyun Kim
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, 134 Sinchon-dong, Seodaemun-gu, Seoul 03722, Republic of Korea.
| | - Joo Yun Won
- Clinical & Translational Research Institute, Anymedi INC., Seoul, South Korea
| | - Seong Yi
- Spine & Spinal Cord Institute, Department of Neurosurgery, College of Medicine, Yonsei University, 134 Sinchon-dong, Seodaemun-gu, Seoul 03722, Republic of Korea.
| |
Collapse
|
3
|
Xu B, Shimauchi-Ohtaki H, Yoshimoto Y, Sadakata T, Ishizaki Y. Transplanted human iPSC-derived vascular endothelial cells promote functional recovery by recruitment of regulatory T cells to ischemic white matter in the brain. J Neuroinflammation 2023; 20:11. [PMID: 36650518 PMCID: PMC9847196 DOI: 10.1186/s12974-023-02694-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 01/06/2023] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Ischemic stroke in white matter of the brain induces not only demyelination, but also neuroinflammation. Peripheral T lymphocytes, especially regulatory T cells (Tregs), are known to infiltrate into ischemic brain and play a crucial role in modulation of inflammatory response there. We previously reported that transplantation of vascular endothelial cells generated from human induced pluripotent stem cells (iVECs) ameliorated white matter infarct. The aim of this study is to investigate contribution of the immune system, especially Tregs, to the mechanism whereby iVEC transplantation ameliorates white matter infarct. METHODS iVECs and human Tregs were transplanted into the site of white matter lesion seven days after induction of ischemia. The egress of T lymphocytes from lymph nodes was sequestered by treating the animals with fingolimod (FTY720). The infarct size was evaluated by magnetic resonance imaging. Immunohistochemistry was performed to detect the activated microglia and macrophages, T cells, Tregs, and oligodendrocyte lineage cells. Remyelination was examined by Luxol fast blue staining. RESULTS iVEC transplantation reduced ED-1+ inflammatory cells and CD4+ T cells, while increased Tregs in the white matter infarct. Treatment of the animals with FTY720 suppressed neuroinflammation and reduced the number of both CD4+ T cells and Tregs in the lesion, suggesting the importance of infiltration of these peripheral immune cells into the lesion in aggravation of neuroinflammation. Suppression of neuroinflammation by FTY720 per se, however, did not promote remyelination in the infarct. FTY720 treatment negated the increase in the number of Tregs by iVEC transplantation in the infarct, and attenuated remyelination promoted by transplanted iVECs, while it did not affect the number of oligodendrocyte lineage cells increased by iVEC transplantation. Transplantation of Tregs together with iVECs into FTY720-treated ischemic white matter did not affect the number of oligodendrocyte lineage cells, while it remarkably promoted myelin regeneration. CONCLUSIONS iVEC transplantation suppresses neuroinflammation, but suppression of neuroinflammation per se does not promote remyelination. Recruitment of Tregs by transplanted iVECs contributes significantly to promotion of remyelination in the injured white matter.
Collapse
Affiliation(s)
- Bin Xu
- grid.256642.10000 0000 9269 4097Department of Molecular and Cellular Neurobiology, Gunma University Graduate School of Medicine, Maebashi, 3-39-22 Showa-Machi, Maebashi, Gunma 371-8511 Japan ,grid.452661.20000 0004 1803 6319Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang China
| | - Hiroya Shimauchi-Ohtaki
- grid.256642.10000 0000 9269 4097Department of Neurosurgery, Gunma University Graduate School of Medicine, Maebashi, Gunma Japan
| | - Yuhei Yoshimoto
- grid.256642.10000 0000 9269 4097Department of Neurosurgery, Gunma University Graduate School of Medicine, Maebashi, Gunma Japan
| | - Tetsushi Sadakata
- grid.256642.10000 0000 9269 4097Education and Research Support Center, Gunma University Graduate School of Medicine, Maebashi, Gunma Japan
| | - Yasuki Ishizaki
- grid.256642.10000 0000 9269 4097Department of Molecular and Cellular Neurobiology, Gunma University Graduate School of Medicine, Maebashi, 3-39-22 Showa-Machi, Maebashi, Gunma 371-8511 Japan
| |
Collapse
|
4
|
Reciprocal Interactions between Oligodendrocyte Precursor Cells and the Neurovascular Unit in Health and Disease. Cells 2022; 11:cells11121954. [PMID: 35741083 PMCID: PMC9221698 DOI: 10.3390/cells11121954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/02/2022] [Accepted: 06/14/2022] [Indexed: 12/04/2022] Open
Abstract
Oligodendrocyte precursor cells (OPCs) are mostly known for their capability to differentiate into oligodendrocytes and myelinate axons. However, they have been observed to frequently interact with cells of the neurovascular unit during development, homeostasis, and under pathological conditions. The functional consequences of these interactions are largely unclear, but are increasingly studied. Although OPCs appear to be a rather homogenous cell population in the central nervous system (CNS), they present with an enormous potential to adapt to their microenvironment. In this review, it is summarized what is known about the various roles of OPC-vascular interactions, and the circumstances under which they have been observed.
Collapse
|
5
|
Abstract
In 2001, the concept of the neurovascular unit was introduced at the Stroke Progress Review Group meeting. The neurovascular unit is an important element of the health and disease status of blood vessels and nerves in the central nervous system. Since then, the neurovascular unit has attracted increasing interest from research teams, who have contributed greatly to the prevention, treatment, and prognosis of stroke and neurodegenerative diseases. However, additional research is needed to establish an efficient, low-cost, and low-energy in vitro model of the neurovascular unit, as well as enable noninvasive observation of neurovascular units in vivo and in vitro. In this review, we first summarize the composition of neurovascular units, then investigate the efficacy of different types of stem cells and cell culture methods in the construction of neurovascular unit models, and finally assess the progress of imaging methods used to observe neurovascular units in recent years and their positive role in the monitoring and investigation of the mechanisms of a variety of central nervous system diseases.
Collapse
Affiliation(s)
- Taiwei Dong
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
| | - Min Li
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
| | - Feng Gao
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
| | - Peifeng Wei
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, China
| | - Jian Wang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Provinve, China
| |
Collapse
|
6
|
Role of extracellular vesicles in neurodegenerative diseases. Prog Neurobiol 2021; 201:102022. [PMID: 33617919 DOI: 10.1016/j.pneurobio.2021.102022] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/27/2020] [Accepted: 02/11/2021] [Indexed: 02/08/2023]
Abstract
Extracellular vesicles (EVs) are heterogeneous cell-derived membranous structures that arise from the endosome system or directly detach from the plasma membrane. In recent years, many advances have been made in the understanding of the clinical definition and pathogenesis of neurodegenerative diseases, but translation into effective treatments is hampered by several factors. Current research indicates that EVs are involved in the pathology of diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), and Huntington's disease (HD). Besides, EVs are also involved in the process of myelin formation, and can also cross the blood-brain barrier to reach the sites of CNS injury. It is suggested that EVs have great potential as a novel therapy for the treatment of neurodegenerative diseases. Here, we reviewed the advances in understanding the role of EVs in neurodegenerative diseases and addressed the critical function of EVs in the CNS. We have also outlined the physiological mechanisms of EVs in myelin regeneration and highlighted the therapeutic potential of EVs in neurodegenerative diseases.
Collapse
|
7
|
Ohashi K, Shibasaki K, Nakazawa H, Kunimasa R, Nagayasu K, Shirakawa H, Kaneko S. Transient Receptor Potential Melastatin 3 Is Functionally Expressed in Oligodendrocyte Precursor Cells and Is Upregulated in Ischemic Demyelinated Lesions. Biol Pharm Bull 2021; 44:181-187. [PMID: 33518671 DOI: 10.1248/bpb.b20-00510] [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] [Indexed: 11/22/2022]
Abstract
Oligodendrocyte precursor cells (OPCs) are glial cells that differentiate into oligodendrocytes and myelinate axons. The number of OPCs is reportedly increased in brain lesions in some demyelinating diseases and during ischemia; however, these cells also secrete cytokines and elicit both protective and deleterious effects in response to brain injury. The mechanism regulating the behaviors of OPCs in physiological and pathological conditions must be elucidated to control these cells and to treat demyelinating diseases. Here, we focused on transient receptor potential melastatin 3 (TRPM3), a Ca2+-permeable channel that is activated by the neurosteroid pregnenolone sulfate (PS) and body temperature. Trpm3+/Pdgfra+ OPCs were detected in the cerebral cortex (CTX) and corpus callosum (CC) of P4 and adult rats by in situ hybridization. Trpm3 expression was detected in primary cultured rat OPCs and was increased by treatment with tumor necrosis factor α (TNFα). Application of PS (30-100 µM) increased the Ca2+ concentration in OPCs and this effect was inhibited by co-treatment with the TRP channel blocker Gd3+ (100 µM) or the TRPM3 inhibitor isosakuranetin (10 µM). Stimulation of TRPM3 with PS (50 µM) did not affect the differentiation or migration of OPCs. The number of Trpm3+ OPCs was markedly increased in demyelinated lesions in an endothelin-1 (ET-1)-induced ischemic rat model. In conclusion, TRPM3 is functionally expressed in OPCs in vivo and in vitro and is upregulated in inflammatory conditions such as ischemic insults and TNFα treatment, implying that TRPM3 is involved in the regulation of specific behaviors of OPCs in pathological conditions.
Collapse
Affiliation(s)
- Kana Ohashi
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Koji Shibasaki
- Division of Neurochemistry, Graduate School of Human Health Science, University of Nagasaki
| | - Hayaki Nakazawa
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Ryotaro Kunimasa
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Kazuki Nagayasu
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Hisashi Shirakawa
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Shuji Kaneko
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University
| |
Collapse
|
8
|
Manukjan N, Ahmed Z, Fulton D, Blankesteijn WM, Foulquier S. A Systematic Review of WNT Signaling in Endothelial Cell Oligodendrocyte Interactions: Potential Relevance to Cerebral Small Vessel Disease. Cells 2020; 9:cells9061545. [PMID: 32630426 PMCID: PMC7349551 DOI: 10.3390/cells9061545] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 06/19/2020] [Accepted: 06/23/2020] [Indexed: 12/21/2022] Open
Abstract
Key pathological features of cerebral small vessel disease (cSVD) include impairment of the blood brain barrier (BBB) and the progression of white matter lesions (WMLs) amongst other structural lesions, leading to the clinical manifestations of cSVD. The function of endothelial cells (ECs) is of major importance to maintain a proper BBB. ECs interact with several cell types to provide structural and functional support to the brain. Oligodendrocytes (OLs) myelinate axons in the central nervous system and are crucial in sustaining the integrity of white matter. The interplay between ECs and OLs and their precursor cells (OPCs) has received limited attention yet seems of relevance for the study of BBB dysfunction and white matter injury in cSVD. Emerging evidence shows a crosstalk between ECs and OPCs/OLs, mediated by signaling through the Wingless and Int-1 (WNT)/β-catenin pathway. As the latter is involved in EC function (e.g., angiogenesis) and oligodendrogenesis, we reviewed the role of WNT/β-catenin signaling for both cell types and performed a systematic search to identify studies describing a WNT-mediated interplay between ECs and OPCs/OLs. Dysregulation of this interaction may limit remyelination of WMLs and render the BBB leaky, thereby initiating a vicious neuroinflammatory cycle. A better understanding of the role of this signaling pathway in EC-OL crosstalk is essential in understanding cSVD development.
Collapse
Affiliation(s)
- Narek Manukjan
- Department of Pharmacology and Toxicology, Maastricht University, PO Box 616, 6200 MD Maastricht, The Netherlands; or (W.M.B.)
- CARIM—School for Cardiovascular Diseases, Maastricht University Medical Center+, PO Box 616, 6200 MD Maastricht, The Netherlands
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK; (Z.A.); (D.F.)
| | - Zubair Ahmed
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK; (Z.A.); (D.F.)
| | - Daniel Fulton
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK; (Z.A.); (D.F.)
| | - W. Matthijs Blankesteijn
- Department of Pharmacology and Toxicology, Maastricht University, PO Box 616, 6200 MD Maastricht, The Netherlands; or (W.M.B.)
- CARIM—School for Cardiovascular Diseases, Maastricht University Medical Center+, PO Box 616, 6200 MD Maastricht, The Netherlands
| | - Sébastien Foulquier
- Department of Pharmacology and Toxicology, Maastricht University, PO Box 616, 6200 MD Maastricht, The Netherlands; or (W.M.B.)
- CARIM—School for Cardiovascular Diseases, Maastricht University Medical Center+, PO Box 616, 6200 MD Maastricht, The Netherlands
- Department of Neurology, MHeNs—School for Mental Health and Neuroscience, Maastricht University Medical Center+, PO Box 616, 6200 MD Maastricht, The Netherlands
- Correspondence: ; Tel.: +31-43-3881409
| |
Collapse
|
9
|
Xu B, Kurachi M, Shimauchi-Ohtaki H, Yoshimoto Y, Ishizaki Y. Transplantation of iPS-derived vascular endothelial cells improves white matter ischemic damage. J Neurochem 2020; 153:759-771. [PMID: 31883380 PMCID: PMC7317957 DOI: 10.1111/jnc.14949] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 12/12/2019] [Accepted: 12/19/2019] [Indexed: 02/06/2023]
Abstract
White matter infarct induces demyelination and brain dysfunction. We previously reported that transplantation of brain microvascular endothelial cells improved the behavioral outcome and promoted remyelination by increasing the number of oligodendrocyte precursor cells in the rat model of white matter infarct. In this study, we investigated the effects of transplantation of vascular endothelial cells generated from human induced pluripotent stem cells (iPSCs) on the rat model of white matter infarct. Seven days after induction of ischemic demyelinating lesion by injection of endothelin‐1 into the internal capsule of a rat brain, iPSC‐derived vascular endothelial cells (iVECs) were transplanted into the site of demyelination. The majority of iVECs transplanted into the internal capsule survived for 14 days after transplantation when traced by immunohistochemistry for a human cytoplasmic protein. iVEC transplantation significantly recovered hind limb rotation angle as compared to human iPSC or rat meningeal cell transplantation when evaluated using footprint test. Fourteen days after iVEC transplantation, the infarct area remarkably decreased as compared to that just before the transplantation when evaluated using magnetic resonance imaging or luxol fast blue staining, and remyelination was promoted dramatically in the infarct when assessed using luxol fast blue staining. Transplantation of iVECs increased the number of oligodendrocyte lineage cells and suppressed the inflammatory response and reactive astrocytogenesis. These results suggest that iVEC transplantation may prove useful in treatment for white matter infarct. ![]()
Collapse
Affiliation(s)
- Bin Xu
- Department of Molecular and Cellular Neurobiology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Masashi Kurachi
- Department of Molecular and Cellular Neurobiology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | | | - Yuhei Yoshimoto
- Department of Neurosurgery, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Yasuki Ishizaki
- Department of Molecular and Cellular Neurobiology, Gunma University Graduate School of Medicine, Maebashi, Japan
| |
Collapse
|
10
|
Shimauchi-Ohtaki H, Kurachi M, Naruse M, Shibasaki K, Sugio S, Matsumoto K, Ema M, Yoshimoto Y, Ishizaki Y. The dynamics of revascularization after white matter infarction monitored in Flt1-tdsRed and Flk1-GFP mice. Neurosci Lett 2018; 692:70-76. [PMID: 30389418 DOI: 10.1016/j.neulet.2018.10.057] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 10/11/2018] [Accepted: 10/30/2018] [Indexed: 12/18/2022]
Abstract
Subcortical white matter infarction causes ischemic demyelination and loss of brain functions, as the result of disturbances of the blood flow. Although angiogenesis is one of the recovery processes after cerebral infarction, the dynamics of revascularization after white matter infarction still remains unclear. We induced white matter infarction in the internal capsule of Flk1-GFP::Flt1-tdsRed double transgenic mice by injection of endothelin-1 (ET-1), a vasoconstrictor peptide, together with N(G)-nitro-L-arginine methyl ester (L-NAME), a nitric oxide synthase inhibitor, and followed the changes in Flk1 and Flt1 expression in the vascular system in the infarct area. Reduction of Flt1-tdsRed-positive blood vessels 1 day after the injection and increase of Flk1-GFP-strongly-positive blood vessels 3 days after the injection were apparent. PDGFRβ-strongly-positive (PDGFRβ+) cells appeared in the infarct area 3 days after the injection and increased their number thereafter. Three days after the injection, most of these cells were in close contact with Flk1-GFP-positive endothelial cells, indicating these cells are bona fide pericytes. Seven days after the injection, the number of PDGFRβ+ cells increased dramatically, and the vast majority of these cells were not in close contact with Flk1-GFP-positive endothelial cells. Taken together, our results suggest revascularization begins early after the ischemic insult, and the emerging pericytes first ensheath blood vessels and then produce fibroblast-like cells not directly associated with blood vessels.
Collapse
Affiliation(s)
- Hiroya Shimauchi-Ohtaki
- Department of Neurosurgery, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan; Department of Molecular and Cellular Neurobiology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Masashi Kurachi
- Department of Molecular and Cellular Neurobiology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Masae Naruse
- Department of Molecular and Cellular Neurobiology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Koji Shibasaki
- Department of Molecular and Cellular Neurobiology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Shouta Sugio
- Department of Molecular and Cellular Neurobiology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Ken Matsumoto
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Masatsugu Ema
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Yuhei Yoshimoto
- Department of Neurosurgery, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Yasuki Ishizaki
- Department of Molecular and Cellular Neurobiology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan.
| |
Collapse
|
11
|
Wu KW, Lv LL, Lei Y, Qian C, Sun FY. Endothelial cells promote excitatory synaptogenesis and improve ischemia-induced motor deficits in neonatal mice. Neurobiol Dis 2018; 121:230-239. [PMID: 30308244 DOI: 10.1016/j.nbd.2018.10.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 09/24/2018] [Accepted: 10/04/2018] [Indexed: 12/11/2022] Open
Abstract
Brain microvascular endothelial cells (BMEC) are highly complex regulatory cells that communicate with other cells in the neurovascular unit. Cerebral ischemic injury is known to produce detectable synaptic dysfunction. This study aims to investigate whether endothelial cells in the brain regulate postnatal synaptic development and to elucidate their role in functional recovery after ischemia. Here, we found that in vivo engraftment of endothelial cells increased synaptic puncta and excitatory postsynaptic currents in layers 2/3 of the motor cortex. This pro-synaptogenic effect was blocked by the depletion of VEGF in the grafted BMEC. The in vitro results showed that BMEC conditioned medium enhanced spine and synapse formation but conditioned medium without VEGF had no such effects. Moreover, under pathological conditions, transplanted endothelial cells were capable of enhancing angiogenesis and synaptogenesis and improved motor function in the ischemic injury model. Collectively, our findings suggest that endothelial cells promote excitatory synaptogenesis via the paracrine factor VEGF during postnatal development and exert repair functions in hypoxia-ischemic neonatal mice. This study highlights the importance of the endothelium-neuron interaction not only in regulating neuronal development but also in maintaining healthy brain function.
Collapse
Affiliation(s)
- Kun-Wei Wu
- Departments of Neurobiology and System Biology for Medicine, Institute for Basic Research on Aging and Medicine, School of Basic Medical Sciences, and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Shanghai Medical College of Fudan University, Shanghai 200032, China; State Key Laboratory of Medical Neurobiology, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Ling-Ling Lv
- Departments of Neurobiology and System Biology for Medicine, Institute for Basic Research on Aging and Medicine, School of Basic Medical Sciences, and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Shanghai Medical College of Fudan University, Shanghai 200032, China; State Key Laboratory of Medical Neurobiology, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Yu Lei
- Departments of Neurobiology and System Biology for Medicine, Institute for Basic Research on Aging and Medicine, School of Basic Medical Sciences, and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Shanghai Medical College of Fudan University, Shanghai 200032, China; State Key Laboratory of Medical Neurobiology, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Cheng Qian
- Departments of Neurobiology and System Biology for Medicine, Institute for Basic Research on Aging and Medicine, School of Basic Medical Sciences, and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Shanghai Medical College of Fudan University, Shanghai 200032, China; State Key Laboratory of Medical Neurobiology, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Feng-Yan Sun
- Departments of Neurobiology and System Biology for Medicine, Institute for Basic Research on Aging and Medicine, School of Basic Medical Sciences, and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Shanghai Medical College of Fudan University, Shanghai 200032, China; State Key Laboratory of Medical Neurobiology, Shanghai Medical College of Fudan University, Shanghai 200032, China.
| |
Collapse
|
12
|
Noumbissi ME, Galasso B, Stins MF. Brain vascular heterogeneity: implications for disease pathogenesis and design of in vitro blood-brain barrier models. Fluids Barriers CNS 2018; 15:12. [PMID: 29688865 PMCID: PMC5911972 DOI: 10.1186/s12987-018-0097-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 04/13/2018] [Indexed: 12/22/2022] Open
Abstract
The vertebrate blood–brain barrier (BBB) is composed of cerebral microvascular endothelial cells (CEC). The BBB acts as a semi-permeable cellular interface that tightly regulates bidirectional molecular transport between blood and the brain parenchyma in order to maintain cerebral homeostasis. The CEC phenotype is regulated by a variety of factors, including cells in its immediate environment and within functional neurovascular units. The cellular composition of the brain parenchyma surrounding the CEC varies between different brain regions; this difference is clearly visible in grey versus white matter. In this review, we discuss evidence for the existence of brain vascular heterogeneity, focusing on differences between the vessels of the grey and white matter. The region-specific differences in the vasculature of the brain are reflective of specific functions of those particular brain areas. This BBB-endothelial heterogeneity may have implications for the course of pathogenesis of cerebrovascular diseases and neurological disorders involving vascular activation and dysfunction. This heterogeneity should be taken into account when developing BBB-neuro-disease models representative of specific brain areas.
Collapse
Affiliation(s)
- Midrelle E Noumbissi
- Malaria Research Institute, Dept. Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, SPH East 4135, Baltimore, MD, 21205, USA
| | - Bianca Galasso
- Malaria Research Institute, Dept. Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, SPH East 4135, Baltimore, MD, 21205, USA
| | - Monique F Stins
- Malaria Research Institute, Dept. Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, SPH East 4135, Baltimore, MD, 21205, USA.
| |
Collapse
|
13
|
Osawa S, Kurachi M, Yamamoto H, Yoshimoto Y, Ishizaki Y. Fibronectin on extracellular vesicles from microvascular endothelial cells is involved in the vesicle uptake into oligodendrocyte precursor cells. Biochem Biophys Res Commun 2017; 488:232-238. [PMID: 28499870 DOI: 10.1016/j.bbrc.2017.05.049] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 05/08/2017] [Indexed: 11/25/2022]
Abstract
We previously reported transplantation of brain microvascular endothelial cells (MVECs) into cerebral white matter infarction model improved the animal's behavioral outcome by increasing the number of oligodendrocyte precursor cells (OPCs). We also revealed extracellular vesicles (EVs) derived from MVECs promoted survival and proliferation of OPCs in vitro. In this study, we investigated the mechanism how EVs derived from MVECs contribute to OPC survival and proliferation. Protein mass spectrometry and enzyme-linked immunosorbent assay revealed fibronectin was abundant on the surface of EVs from MVECs. As fibronectin has been reported to promote OPC survival and proliferation via integrin signaling pathway, we blocked the binding between fibronectin and integrins using RGD sequence mimics. Blocking the binding, however, did not attenuate the survival and proliferation promoting effect of EVs on OPCs. Flow cytometric and imaging analyses revealed fibronectin on EVs mediates their internalization into OPCs by its binding to heparan sulfate proteoglycan on OPCs. OPC survival and proliferation promoted by EVs were attenuated by blocking the internalization of EVs into OPCs. These lines of evidence suggest that fibronectin on EVs mediates their internalization into OPCs, and the cargo of EVs promotes survival and proliferation of OPCs, independent of integrin signaling pathway.
Collapse
Affiliation(s)
- Sho Osawa
- Department of Neurosurgery, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan; Department of Molecular and Cellular Neurobiology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Masashi Kurachi
- Department of Molecular and Cellular Neurobiology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Hanako Yamamoto
- Department of Molecular and Cellular Neurobiology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Yuhei Yoshimoto
- Department of Neurosurgery, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Yasuki Ishizaki
- Department of Molecular and Cellular Neurobiology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan.
| |
Collapse
|
14
|
Wu KW, Kou ZW, Mo JL, Deng XX, Sun FY. Neurovascular coupling protects neurons against hypoxic injury via inhibition of potassium currents by generation of nitric oxide in direct neuron and endothelium cocultures. Neuroscience 2016; 334:275-282. [PMID: 27531855 DOI: 10.1016/j.neuroscience.2016.08.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 07/15/2016] [Accepted: 08/07/2016] [Indexed: 12/15/2022]
Abstract
This study examined the effect of neuron-endothelial coupling on the survival of neurons after ischemia and the possible mechanism underlying that effect. Whole-cell patch-clamp experiments were performed on cortical neurons cultured alone or directly cocultured with brain microvascular endothelial cells (BMEC). Propidium iodide (PI) and NeuN staining were performed to examine neuronal death following oxygen and glucose deprivation (OGD). We found that the neuronal transient outward potassium currents (IA) decreased in the coculture system, whereas the outward delayed-rectifier potassium currents (IK) did not. Sodium nitroprusside, a NO donor, enhanced BMEC-induced IA inhibition and nitro-l-arginine methylester, a NOS inhibitor, partially prevented this inhibition. Moreover, the neurons directly cocultured with BMEC showed more resistance to OGD-induced injury compared with the neurons cultured alone, and that neuroprotective effect was abolished by treatment with NS5806, an activator of the IA. These results indicate that vascular endothelial cells assist neurons to prevent hypoxic injury via inhibiting neuronal IA by production of NO in the direct neuron-BMEC coculture system. These results further provide direct evidence of functional coupling between neurons and vascular endothelial cells. This study clearly demonstrates that vascular endothelial cells play beneficial roles in the pathophysiological processes of neurons after hypoxic injury, suggesting that the improvement of neurovascular coupling or functional remodeling may become an important therapeutic target for preventing brain injury.
Collapse
Affiliation(s)
- Kun-Wei Wu
- Institute of Biomedical Sciences and Department of Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, PR China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Research Center on Aging and Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, PR China
| | - Zeng-Wei Kou
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Research Center on Aging and Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, PR China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, PR China
| | - Jia-Lin Mo
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Research Center on Aging and Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, PR China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, PR China
| | - Xu-Xu Deng
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Research Center on Aging and Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, PR China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, PR China
| | - Feng-Yan Sun
- Institute of Biomedical Sciences and Department of Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, PR China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Research Center on Aging and Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, PR China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, PR China.
| |
Collapse
|
15
|
Kurachi M, Mikuni M, Ishizaki Y. Extracellular Vesicles from Vascular Endothelial Cells Promote Survival, Proliferation and Motility of Oligodendrocyte Precursor Cells. PLoS One 2016; 11:e0159158. [PMID: 27403742 PMCID: PMC4942096 DOI: 10.1371/journal.pone.0159158] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 06/28/2016] [Indexed: 01/02/2023] Open
Abstract
We previously examined the effect of brain microvascular endothelial cell (MVEC) transplantation on rat white matter infarction, and found that MVEC transplantation promoted remyelination of demyelinated axons in the infarct region and reduced apoptotic death of oligodendrocyte precursor cells (OPCs). We also found that the conditioned medium (CM) from cultured MVECs inhibited apoptosis of cultured OPCs. In this study, we examined contribution of extracellular vesicles (EVs) contained in the CM to its inhibitory effect on OPC apoptosis. Removal of EVs from the CM by ultracentrifugation reduced its inhibitory effect on OPC apoptosis. To confirm whether EVs derived from MVECs are taken up by cultured OPCs, we labeled EVs with PKH67, a fluorescent dye, and added them to OPC cultures. Many vesicular structures labeled with PKH67 were found within OPCs immediately after their addition. Next we examined the effect of MVEC-derived EVs on OPC behaviors. After 2 days in culture with EVs, there was significantly less pyknotic and more BrdU-positive OPCs when compared to control. We also examined the effect of EVs on motility of OPCs. OPCs migrated longer in the presence of EVs when compared to control. To examine whether these effects on cultured OPCs are shared by EVs from endothelial cells, we prepared EVs from conditioned media of several types of endothelial cells, and tested their effects on cultured OPCs. EVs from all types of endothelial cells we examined reduced apoptosis of OPCs and promoted their motility. Identification of the molecules contained in EVs from endothelial cells may prove helpful for establishment of effective therapies for demyelinating diseases.
Collapse
Affiliation(s)
- Masashi Kurachi
- Department of Molecular and Cellular Neurobiology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Masahiko Mikuni
- Department of Psychiatry and Neuroscience, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Yasuki Ishizaki
- Department of Molecular and Cellular Neurobiology, Gunma University Graduate School of Medicine, Maebashi, Japan
- * E-mail:
| |
Collapse
|