1
|
Kang S, Lee DA, Lee JW, Lee HJ, Park KM. White matter changes in patients with narcolepsy type 2: Peak width of skeletonized mean diffusivity study. Sleep Med 2025; 129:14-19. [PMID: 39970700 DOI: 10.1016/j.sleep.2025.02.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 02/10/2025] [Accepted: 02/14/2025] [Indexed: 02/21/2025]
Abstract
OBJECTIVES This study aimed to investigate white matter (WM) microstructural alterations in patients with narcolepsy type 2 (NT2) using Peak Width of Skeletonized Mean Diffusivity (PSMD), a novel imaging marker associated with small vessel disease (SVD). The study compared PSMD metrics between patients with NT2 and healthy controls to investigate structural disruptions and their implications for NT2 pathophysiology. METHODS A total of 42 participants were enrolled, including 20 patients with newly diagnosed NT2 and 22 healthy controls. Diffusion tensor imaging (DTI) was performed using a 3 T MRI scanner. PSMD was calculated using a multi-step process involving preprocessing, skeletonization, application of a custom mask, and histogram analysis with the FSL program. PSMD values were compared between patients with NT2 and healthy controls, and correlation analyses were conducted to examine associations between PSMD and clinical variables. RESULTS Patients with NT2 exhibited significantly higher PSMD compared to healthy controls (2.172 × 10-4 mm2/s vs. 2.031 × 10-4 mm2/s, p = 0.011). PSMD also positively correlated with age in both patients with NT2 (r = 0.608, p = 0.004) and healthy controls (r = 0.696, p < 0.001). CONCLUSION Patients with NT2 demonstrate increased PSMD, indicating WM microstructural changes potentially linked to SVD. These findings highlight the utility of PSMD as a sensitive neuroimaging marker for detecting WM alterations in sleep disorders. Further studies are needed to validate these results and investigate the underlying mechanisms of WM changes in NT2.
Collapse
Affiliation(s)
- Sujin Kang
- Asan Medical Center, Seoul, Republic of Korea
| | - Dong Ah Lee
- Department of Neurology, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - Jun Won Lee
- Department of Neurology, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - Ho-Joon Lee
- Department of Radiology, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - Kang Min Park
- Department of Neurology, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea.
| |
Collapse
|
2
|
Ruscher K, Michalettos G, Abu Hamdeh S, Clausen F, Nolan AL, Flygt J, Özen I, Marklund N. Persistent increase of Nogo-A-positive cells and dynamic reduction in oligodendroglia lineage cells in white matter regions following experimental and clinical traumatic brain injury. J Neuropathol Exp Neurol 2025; 84:423-435. [PMID: 40085014 PMCID: PMC12012378 DOI: 10.1093/jnen/nlaf017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2025] Open
Abstract
White matter (WM) disruption and atrophy is a consequence of traumatic brain injury (TBI) and contributes to persisting cognitive impairment. An increased expression of the myelin-associated axonal outgrowth inhibitor Nogo-A and oligodendrocyte pathology might be negatively associated with postinjury WM changes. Here, we analyzed brain tissue from severe TBI patients, obtained by surgical decompression in the early postinjury phase and postmortem brain tissue of long-term TBI survivors and observed an increased number of Nogo-A+ cells in WM tracts such as the corpus callosum (CC). Likewise, the number of Nogo-A+ cells in the CC was increased from day 7 postinjury to 6 months postinjury (mpi) following central fluid percussion injury (cFPI) in mice. In addition, the number of Olig2+ cells in the CC and capsula externa remained constant, while the numbers of Olig2+/CC1+ and GST-π+ mature oligodendrocytes declined throughout the observation time of 18 months. A significantly lower number of Olig2+/CC1+ cells was found in cFPI mice compared to controls at 18 mpi. Persistent vulnerability of oligodendrocytes in combination with dynamic alterations of Nogo-A expression may have implications for the WM atrophy and insufficient recovery observed after TBI.
Collapse
Affiliation(s)
- Karsten Ruscher
- Department of Clinical Sciences Lund, Neurosurgery, Lund University, Lund, Sweden
- Department of Clinical Sciences, Division of Neurosurgery, Laboratory for Experimental Brain Research, Lund University, Lund, Sweden
| | - Georgios Michalettos
- Department of Clinical Sciences Lund, Neurosurgery, Lund University, Lund, Sweden
| | - Sami Abu Hamdeh
- Department of Medical Sciences, Section of Neurosurgery, Uppsala University, Uppsala, Sweden
| | - Fredrik Clausen
- Department of Medical Sciences, Section of Neurosurgery, Uppsala University, Uppsala, Sweden
| | - Amber L Nolan
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
| | - Johanna Flygt
- Department of Medical Sciences, Section of Neurosurgery, Uppsala University, Uppsala, Sweden
| | - Ilknur Özen
- Department of Clinical Sciences Lund, Neurosurgery, Lund University, Lund, Sweden
| | - Niklas Marklund
- Department of Clinical Sciences Lund, Neurosurgery, Lund University, Lund, Sweden
- Department of Clinical Sciences, Neurosurgery, Lund University, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
3
|
Xue H, Ding Z, Chen X, Yang X, Jia Y, Zhao P, Wu Z. Dexmedetomidine Improves Long-term Neurological Outcomes by Promoting Oligodendrocyte Genesis and Myelination in Neonatal Rats Following Hypoxic-ischemic Brain Injury. Mol Neurobiol 2025; 62:4866-4880. [PMID: 39496877 DOI: 10.1007/s12035-024-04564-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 10/18/2024] [Indexed: 11/06/2024]
Abstract
Neonatal hypoxic-ischemic brain injury (HIBI) can lead to white matter damage, which significantly contributes to cognitive dysfunction, emotional disorders, and sensorimotor impairments. Although dexmedetomidine enhances neurobehavioral outcomes, its impact on oligodendrocyte genesis and myelination following hypoxic-ischemic events, as well as the underlying mechanisms, remain poorly understood. Dexmedetomidine was administered 15 min post-HIBI. We assessed neurobehavioral deficits using various tests: surface righting, negative geotaxis, forelimb grip strength, cliff avoidance, sensory reflexes, novel object recognition, T-maze, and three-chamber social interaction. We also investigated the relationship between myelination and neurobehavioral outcomes. Measurements included oligodendrocyte precursor cell (OPC) proliferation and survival 24 h post-injury, early myelination, and oligodendrocyte differentiation by postnatal day 14. Furthermore, we evaluated microglial activation towards the M2 phenotype and the extent of neuroinflammation during the acute phase. Dexmedetomidine significantly ameliorated long-term neurological deficits caused by HIBI. Pearson linear regression analysis revealed a strong correlation between long-term neurological outcomes and myelin maturity. The treatment notably mitigated the long-term deterioration of myelin formation and maturation following HIBI. This protective effect was primarily due to enhanced OPC proliferation and survival post-HIBI during the acute phase and, to a lesser extent, to the modulation of microglial activity towards the M2 phenotype and a reduction in neuroinflammation. Dexmedetomidine offers substantial protection against long-term neurobehavioral disabilities induced by HIBI, primarily by revitalizing the impaired survival and maturation of oligodendrocyte progenitor cells and promoting myelination.
Collapse
Affiliation(s)
- Hang Xue
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Zixuan Ding
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Xiaoyan Chen
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Xu Yang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Yufei Jia
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Ping Zhao
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Ziyi Wu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| |
Collapse
|
4
|
Shanks MJ, Byblow WD. Corticomotor pathway function and recovery after stroke: a look back and a way forward. J Physiol 2025; 603:651-662. [PMID: 38814805 PMCID: PMC11782909 DOI: 10.1113/jp285562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 05/15/2024] [Indexed: 06/01/2024] Open
Abstract
Stroke is a leading cause of adult disability that results in motor deficits and reduced independence. Regaining independence relies on motor recovery, particularly regaining function of the hand and arm. This review presents evidence from human studies that have used transcranial magnetic stimulation (TMS) to identify neurophysiological mechanisms underlying upper limb motor recovery early after stroke. TMS studies undertaken at the subacute stage after stroke have identified several neurophysiological factors that can drive motor impairment, including membrane excitability, the recruitment of corticomotor neurons, and glutamatergic and GABAergic neurotransmission. However, the inherent variability and subsequent poor reliability of measures derived from motor evoked potentials (MEPs) limit the use of TMS for prognosis at the individual patient level. Currently, prediction tools that provide the most accurate information about upper limb motor outcomes for individual patients early after stroke combine clinical measures with a simple neurophysiological biomarker based on MEP presence or absence, i.e. MEP status. Here, we propose a new compositional framework to examine MEPs across several upper limb muscles within a threshold matrix. The matrix can provide a more comprehensive view of corticomotor function and recovery after stroke by quantifying the evolution of subthreshold and suprathreshold MEPs through compositional analyses. Our contention is that subthreshold responses might be the most sensitive to reduced output of corticomotor neurons, desynchronized firing of the remaining neurons, and myelination processes that occur early after stroke. Quantifying subthreshold responses might provide new insights into post-stroke neurophysiology and improve the accuracy of prediction of upper limb motor outcomes.
Collapse
Affiliation(s)
- Maxine J. Shanks
- Department of Exercise SciencesUniversity of AucklandAucklandNew Zealand
- Centre for Brain ResearchUniversity of AucklandAucklandNew Zealand
| | - Winston D. Byblow
- Department of Exercise SciencesUniversity of AucklandAucklandNew Zealand
- Centre for Brain ResearchUniversity of AucklandAucklandNew Zealand
| |
Collapse
|
5
|
Zheng L, Wang X, Li P, Hao Y, Zhang H, Chen H, Zheng Z. White Matter Injury in Central Nervous System Disorders. Neuropsychiatr Dis Treat 2025; 21:107-114. [PMID: 39877856 PMCID: PMC11774249 DOI: 10.2147/ndt.s498660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 01/13/2025] [Indexed: 01/31/2025] Open
Abstract
As the aging process accelerates and living conditions improve, central nervous system (CNS) diseases have become a major public health problem. Diseases of the CNS cause not only gray matter damage, which is primarily characterized by the loss of neurons, but also white matter damage. However, most previous studies have focused on grey matter injury (GMI), with fewer studies on white matter injury (WMI). In this article, we will briefly describe the structure and function of white matter, summarize the pathological changes of WMI, and focus on the molecular mechanisms and therapeutic research advances in WMI after ischemic stroke, cerebral hemorrhage, Alzheimer's disease, and multiple sclerosis diseases.
Collapse
Affiliation(s)
- Ling Zheng
- Department of Rehabilitation Medicine, The Affiliated Taian City Central Hospital of Qingdao University, Taian, 271000, People’s Republic of China
| | - Xiaoyu Wang
- Department of Rehabilitation Medicine, The Affiliated Taian City Central Hospital of Qingdao University, Taian, 271000, People’s Republic of China
| | - Peng Li
- Department of Rehabilitation Medicine, The Affiliated Taian City Central Hospital of Qingdao University, Taian, 271000, People’s Republic of China
| | - Yuwen Hao
- Department of Rehabilitation Medicine, The Affiliated Taian City Central Hospital of Qingdao University, Taian, 271000, People’s Republic of China
| | - Hao Zhang
- Department of Rehabilitation Medicine, The Affiliated Taian City Central Hospital of Qingdao University, Taian, 271000, People’s Republic of China
| | - Haoyue Chen
- Department of Rehabilitation Medicine, The Affiliated Taian City Central Hospital of Qingdao University, Taian, 271000, People’s Republic of China
| | - Zuncheng Zheng
- Department of Rehabilitation Medicine, The Affiliated Taian City Central Hospital of Qingdao University, Taian, 271000, People’s Republic of China
| |
Collapse
|
6
|
Gong XR, You XR, Guo MR, Ding XY, Ma BX. Exploring the mechanism of Pujin oral liquid in the treatment of preterm white matter injury using network pharmacology and molecular docking. Medicine (Baltimore) 2025; 104:e40799. [PMID: 40184095 PMCID: PMC11709195 DOI: 10.1097/md.0000000000040799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 01/04/2024] [Accepted: 11/14/2024] [Indexed: 04/05/2025] Open
Abstract
We aimed to elucidate the pharmacological mechanisms of Pujin oral liquid in treating preterm white matter injury (PWMI). The Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform was used to identify Pujin oral liquid's active ingredients and predict their targets. The known targets related to treating PWMI were identified from the GeneCards, Online Mendelian Inheritance in Man, DisGeNet, PharmGKB, and CTD databases. A drug-disease intersecting protein-protein interaction network using a STRING database was built; gene ontology function and Kyoto Encyclopedia of Genes and Genomes signaling pathway enrichment analyses were performed on common target genes using the Metascape database. Molecular docking of the active ingredients and key targets was validated using the AutoDock Vina software. In total, 470 Pujin oral liquid targets and 13,290 disease targets were screened from multiple databases, and Venn analysis identified 407 common targets. Protein-protein interaction analysis showed that Pujin oral liquid may impact SRC, MAPK3, MAPK1, TP53, STAT3, AKT1, PIK3R1, JUN, RELA, CTNNB1, and ESR1. Moreover, gene ontology functional analysis revealed processes such as the response to inorganic substances, cellular response to organic cyclic compounds, response to xenobiotic stimuli, regulation of system processes, and protein phosphorylation. The main signaling pathways were neuroactive ligand-receptor interaction and the cGMP-PKG, JAK-STAT, and cAMP signaling pathways. Molecular docking showed that the active ingredients' small molecules bond strongly to target proteins. The therapeutic effect of Pujin oral liquid on PWMI is multifaceted, involving multiple targets and pathways. Its clinical application in treating preterm white matter injuries is promising.
Collapse
Affiliation(s)
- Xing-Ruo Gong
- Department of Pediatrics, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiao-Rui You
- Hebei University of Chinese Medicine, Shijiazhuang, Hebei Province, China
| | - Mei-Ran Guo
- Hebei University of Chinese Medicine, Shijiazhuang, Hebei Province, China
| | - Xue-Ying Ding
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan Province, China
| | - Bing-Xiang Ma
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan Province, China
| |
Collapse
|
7
|
Zhang N, Peng K, Guo JX, Liu Q, Xiao AL, Jing H. Microstructural brain abnormalities and associated neurocognitive dysfunction in obstructive sleep apnea: a pilot study with diffusion kurtosis imaging. J Clin Sleep Med 2024; 20:1571-1578. [PMID: 38656791 PMCID: PMC11446125 DOI: 10.5664/jcsm.11184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 04/10/2024] [Accepted: 04/12/2024] [Indexed: 04/26/2024]
Abstract
STUDY OBJECTIVES We assessed possible brain abnormalities in adult patients with moderate and severe obstructive sleep apnea using the mean kurtosis (MK) from diffusion kurtosis imaging and analyzed the correlation between MK and cognitive function. METHODS A total of 30 patients with moderate or severe obstructive sleep apnea and 30 healthy controls evaluated by the Montreal Cognitive Assessment scale were enrolled. All participants underwent diffusion kurtosis imaging and 3-dimensional T1-weighted imaging on a 3.0T magnetic resonance scanner. The MK values of gray and white matter brain regions were compared. Partial correlation analysis was used to analyze the correlation between respiratory sleep parameters/cognitive score and MK values in different brain regions. RESULTS Compared with the healthy controls, the MK of 20 brain regions (13 after false discovery rate correction) and cognitive scores in the obstructive sleep apnea group were significantly lower. In the obstructive sleep apnea group, apnea-hypopnea index was negatively correlated with the MK in the white matter of the right occipital lobe; lowest oxygen saturation was positively correlated with the MK in the bilateral parietal, precentral, and right postcentral cortex; total score on the Montreal Cognitive Assessment scale was positively correlated with MK in the left hippocampus; language function was positively correlated with MK in the white matter of the left parietal lobe; and delayed recall was positively correlated with the MK in right insula cortex and bilateral cingulate. After false discovery rate correction, only the correlations of lowest oxygen saturation with right precentral gyrus cortex and bilateral parietal cortex were significant. CONCLUSIONS MK values of diffusion kurtosis imaging may provide valuable information in assessing the neurological impacts of obstructive sleep apnea. CITATION Zhang N, Peng K, Guo J-X, Liu Q, Xiao A-L, Jing H. Microstructural brain abnormalities and associated neurocognitive dysfunction in obstructive sleep apnea: a pilot study with diffusion kurtosis imaging. J Clin Sleep Med. 2024;20(10):1571-1578.
Collapse
Affiliation(s)
- Ning Zhang
- Department of Radiology, The Sixth Hospital of Shanxi Medical University (General Hospital of TISCO), Taiyuan, People’s Republic of China
| | - Kun Peng
- Department of Radiology, The Sixth Hospital of Shanxi Medical University (General Hospital of TISCO), Taiyuan, People’s Republic of China
| | - Jin-Xia Guo
- GE Healthcare, Beijing, People’s Republic of China
| | - Qing Liu
- College of Medical Imaging, Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Ai-Lian Xiao
- Department of Respiratory and Critical Care Medicine, The Sixth Hospital of Shanxi Medical University (General Hospital of TISCO), Taiyuan, People’s Republic of China
| | - Hui Jing
- Department of Radiology, The Sixth Hospital of Shanxi Medical University (General Hospital of TISCO), Taiyuan, People’s Republic of China
| |
Collapse
|
8
|
Sun E, Lu S, Yang C, Li Z, Qian Y, Chen Y, Chen S, Ma X, Deng Y, Shan X, Chen B. Hypothermia protects the integrity of corticospinal tracts and alleviates mitochondria injury after intracerebral hemorrhage in mice. Exp Neurol 2024; 377:114803. [PMID: 38679281 DOI: 10.1016/j.expneurol.2024.114803] [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: 02/19/2024] [Revised: 04/08/2024] [Accepted: 04/25/2024] [Indexed: 05/01/2024]
Abstract
Disruption of corticospinal tracts (CST) is a leading factor for motor impairments following intracerebral hemorrhage (ICH) in the striatum. Previous studies have shown that therapeutic hypothermia (HT) improves outcomes of ICH patients. However, whether HT has a direct protection effect on the CST integrity and the underlying mechanisms remain largely unknown. In this study, we employed a chemogenetics approach to selectively activate bilateral warm-sensitive neurons in the preoptic areas to induce a hypothermia-like state. We then assessed effects of HT treatment on the integrity of CST and motor functional recovery after ICH. Our results showed that HT treatment significantly alleviated axonal degeneration around the hematoma and the CST axons at remote midbrain region, ultimately promoted skilled motor function recovery. Anterograde and retrograde tracing revealed that HT treatment protected the integrity of the CST over an extended period. Mechanistically, HT treatment prevented mitochondrial swelling in degenerated axons around the hematoma, alleviated mitochondrial impairment by reducing mitochondrial ROS accumulation and improving mitochondrial membrane potential in primarily cultured cortical neurons with oxyhemoglobin treatment. Serving as a proof of principle, our study provided novel insights into the application of HT to improve functional recovery after ICH.
Collapse
Affiliation(s)
- Eryi Sun
- Department of Neurosurgery, The Affiliated People's Hospital of Jiangsu University, Zhenjiang 212002, China
| | - Siyuan Lu
- Department of Radiological, The Affiliated People's Hospital of Jiangsu University, Zhenjiang 212002, China
| | - Chuanyan Yang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Zheng Li
- Department of Neurosurgery, The Affiliated People's Hospital of Jiangsu University, Zhenjiang 212002, China
| | - Yu Qian
- Department of Neurosurgery, The Affiliated People's Hospital of Jiangsu University, Zhenjiang 212002, China
| | - Yue Chen
- Chengdu Bio-HT Company Limited, Chengdu 610000, Sichuan, China
| | - Siyuan Chen
- Department of Neurology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang 212002, China
| | - Xiaodong Ma
- Department of Anesthesiology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang 212002, China
| | - Yan Deng
- Department of Anesthesiology, West China Hospital, Sichuan University, Sichuan, China
| | - Xiuhong Shan
- Department of Radiological, The Affiliated People's Hospital of Jiangsu University, Zhenjiang 212002, China
| | - Bo Chen
- Department of Neurosurgery, The Affiliated People's Hospital of Jiangsu University, Zhenjiang 212002, China.
| |
Collapse
|
9
|
Huang WT, Chen XJ, Lin YK, Shi JF, Li H, Wu HD, Jiang RL, Chen S, Wang X, Tan XX, Chen KY, Wang P. FGF17 protects cerebral ischemia reperfusion-induced blood-brain barrier disruption via FGF receptor 3-mediated PI3K/AKT signaling pathway. Eur J Pharmacol 2024; 971:176521. [PMID: 38522639 DOI: 10.1016/j.ejphar.2024.176521] [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: 06/27/2023] [Revised: 03/07/2024] [Accepted: 03/21/2024] [Indexed: 03/26/2024]
Abstract
Maintaining blood-brain barrier (BBB) integrity is critical components of therapeutic approach for ischemic stroke. Fibroblast growth factor 17 (FGF17), a member of FGF8 superfamily, exhibits the strongest expression throughout the wall of all major arteries during development. However, its molecular action and potential protective role on brain endothelial cells after stroke remains unclear. Here, we observed reduced levels of FGF17 in the serum of patients with ischemic stroke, as well as in the brains of mice subjected to middle cerebral artery occlusion (MCAO) injury and oxygen-glucose deprivation/reoxygenation (OGD/R)-induced brain microvascular endothelial cells (bEnd.3) cells. Moreover, treatment with exogenous recombinant human FGF17 (rhFGF17) decreased infarct volume, improved neurological deficits, reduced Evans Blue leakage and upregulated the expression of tight junctions in MCAO-injured mice. Meanwhile, rhFGF17 increased cell viability, enhanced trans-endothelial electrical resistance, reduced sodium fluorescein leakage, and alleviated reactive oxygen species (ROS) generation in OGD/R-induced bEnd.3 cells. Mechanistically, the treatment with rhFGF17 resulted in nuclear factor erythroid 2-related factor 2 (Nrf2) nuclear accumulation and upregulation of heme oxygenase-1 (HO-1) expression. Additionally, based on in-vivo and in-vitro research, rhFGF17 exerted protective effects against ischemia/reperfusion (I/R) -induced BBB disruption and endothelial cell apoptosis through the activation of the FGF receptor 3/PI3K/AKT signaling pathway. Overall, our findings indicated that FGF17 may hold promise as a novel therapeutic strategy for ischemic stroke patients.
Collapse
Affiliation(s)
- Wen-Ting Huang
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Xiong-Jian Chen
- Department of Pharmacy, Wenzhou Central Hospital, Wenzhou, 325099, China
| | - Yu-Kai Lin
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Jun-Feng Shi
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Hong Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Hao-Di Wu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Ruo-Lin Jiang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Shuai Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Xue Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Xian-Xi Tan
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China.
| | - Ke-Yang Chen
- Department of Neurology, The Second Affiliated Hospital and Yuying Children' Hospital of Wenzhou Medical University, Wenzhou, 325027, China.
| | - Peng Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
10
|
Wu S, Tabassum S, Payne CT, Hu H, Gusdon AM, Choi HA, Ren XS. Updates of the role of B-cells in ischemic stroke. Front Cell Neurosci 2024; 18:1340756. [PMID: 38550918 PMCID: PMC10972894 DOI: 10.3389/fncel.2024.1340756] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 02/27/2024] [Indexed: 10/11/2024] Open
Abstract
Ischemic stroke is a major disease causing death and disability in the elderly and is one of the major diseases that seriously threaten human health and cause a great economic burden. In the early stage of ischemic stroke, neuronal structure is destroyed, resulting in death or damage, and the release of a variety of damage-associated pattern molecules induces an increase in neuroglial activation, peripheral immune response, and secretion of inflammatory mediators, which further exacerbates the damage to the blood-brain barrier, exacerbates cerebral edema, and microcirculatory impairment, triggering secondary brain injuries. After the acute phase of stroke, various immune cells initiate a protective effect, which is released step by step and contributes to the repair of neuronal cells through phenotypic changes. In addition, ischemic stroke induces Central Nervous System (CNS) immunosuppression, and the interaction between the two influences the outcome of stroke. Therefore, modulating the immune response of the CNS to reduce the inflammatory response and immune damage during stroke is important for the protection of brain function and long-term recovery after stroke, and modulating the immune function of the CNS is expected to be a novel therapeutic strategy. However, there are fewer studies on B-cells in brain function protection, which may play a dual role in the stroke process, and the understanding of this cell is still incomplete. We review the existing studies on the mechanisms of the role of B-cells, inflammatory response, and immune response in the development of ischemic stroke and provide a reference for the development of adjuvant therapeutic drugs for ischemic stroke targeting inflammatory injury.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xuefang S. Ren
- Division of Neurocritical Care, Department of Neurosurgery, McGovern School of Medicine, University of Texas Health Science Center, Houston, TX, United States
| |
Collapse
|
11
|
Wang XX, Li GS, Wang KH, Hu XS, Hu Y. Positive effect of microvascular proliferation on functional recovery in experimental cervical spondylotic myelopathy. Front Neurosci 2024; 18:1254600. [PMID: 38510463 PMCID: PMC10951064 DOI: 10.3389/fnins.2024.1254600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 02/09/2024] [Indexed: 03/22/2024] Open
Abstract
Background and purpose Cervical Spondylotic Myelopathy (CSM), the most common cause of spinal cord dysfunction globally, is a degenerative disease that results in non-violent, gradual, and long-lasting compression of the cervical spinal cord. The objective of this study was to investigate whether microvascular proliferation could positively affect neural function recovery in experimental cervical spondylotic myelopathy (CSM). Methods A total of 60 male adult Sprague-Dawley (SD) were randomly divided into four groups: Control (CON), Compression (COM), Angiostasis (AS), and Angiogenesis (A G),with 15 rats in each group. Rats in the AS group received SU5416 to inhibit angiogenesis, while rats in the AG group received Deferoxamine (DFO) to promote angiogenesis. Motor and sensory functions were assessed using the Basso Beattie Bresnahan (BBB) scale and somatosensory evoked potential (SEP) examination. Neuropathological degeneration was evaluated by the number of neurons, Nissl bodies (NB), and the de-myelination of white matter detected by Hematoxylin & Eosin(HE), Toluidine Blue (TB), and Luxol Fast Blue (LFB) staining. Immunohistochemical (IHC) staining was used to observe the Neurovascular Unit (NVU). Results Rats in the CON group exhibited normal locomotor function with full BBB score, normal SEP latency and amplitude. Among the other three groups, the AG group had the highest BBB score and the shortest SEP latency, while the AS group had the lowest BBB score and the most prolonged SEP latency. The SEP amplitude showed an opposite performance to the latency. Compared to the COM and AS groups, the AG group demonstrated significant neuronal restoration in gray matter and axonal remyelination in white matter. DFO promoted microvascular proliferation, especially in gray matter, and improved the survival of neuroglial cells. In contrast, SU-5416 inhibited the viability of neuroglial cells by reducing micro vessels. Conclusion The microvascular status was closely related to NVU remodeling an-d functional recovery. Therefore, proliferation of micro vessels contributed to function -al recovery in experimental CSM, which may be associated with NVU remodeling.
Collapse
Affiliation(s)
- Xu-xiang Wang
- Department of Minimally Invasive Spine Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Guang-sheng Li
- Department of Minimally Invasive Spine Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Department of Orthopaedics and Traumatology, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Kang-heng Wang
- Department of Minimally Invasive Spine Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Department of Orthopaedics and Traumatology, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Xiao-song Hu
- Department of Minimally Invasive Spine Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Orthopedics Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Yong Hu
- Department of Minimally Invasive Spine Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Department of Orthopaedics and Traumatology, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Orthopedics Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
12
|
Visco DB, Manhães-de-Castro R, da Silva MM, Costa-de-Santana BJR, Pereira Dos Santos Junior J, Saavedra LM, de Lemos MDTB, Valdéz-Alarcón JJ, Lagranha CJ, Guzman-Quevedo O, Torner L, Toscano AE. Neonatal kaempferol exposure attenuates impact of cerebral palsy model on neuromotor development, cell proliferation, microglia activation, and antioxidant enzyme expression in the hippocampus of rats. Nutr Neurosci 2024; 27:20-41. [PMID: 36576161 DOI: 10.1080/1028415x.2022.2156034] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
OBJECTIVES This study aims to assess the effect of neonatal treatment with kaempferol on neuromotor development, proliferation of neural precursor cells, the microglia profile, and antioxidant enzyme gene expression in the hippocampus. METHODS A rat model of cerebral palsy was established using perinatal anoxia and sensorimotor restriction of hindlimbs during infancy. Kaempferol (1 mg/ kg) was intraperitoneally administered during the neonatal period. RESULTS Neonatal treatment with kaempferol reduces the impact of the cerebral palsy model on reflex ontogeny and on the maturation of physical features. Impairment of locomotor activity development and motor coordination was found to be attenuated by kaempferol treatment during the neonatal period in rats exposed to cerebral palsy. Neonatal treatment of kaempferol in cerebral palsy rats prevents a substantial reduction in the number of neural precursor cells in the dentate gyrus of the hippocampus, an activated microglia profile, and increased proliferation of microglia in the sub-granular zone and in the granular cell layer. Neonatal treatment with kaempferol increases gene expression of superoxide dismutase and catalase in the hippocampus of rats submitted to the cerebral palsy model. DISCUSSION Kaempferol attenuates the impact of cerebral palsy on neuromotor behavior development, preventing altered hippocampal microglia activation and mitigating impaired cell proliferation in a neurogenic niche in these rats. Neonatal treatment with kaempferol also increases antioxidant defense gene expression in the hippocampus of rats submitted to the cerebral palsy model.
Collapse
Affiliation(s)
- Diego Bulcão Visco
- Laboratory of Neurofunctional, Department of Biological Sciences and Health, Federal University of Amapá, Macapá, Brazil
- Graduate Program in Nutrition (Posnutri), Health Sciences Center, Federal University of Pernambuco, Recife, Brazil
- Studies in Nutrition and Phenotypic Plasticity Unit, Department of Nutrition, Federal University of Pernambuco, Recife, Brazil
| | - Raul Manhães-de-Castro
- Graduate Program in Nutrition (Posnutri), Health Sciences Center, Federal University of Pernambuco, Recife, Brazil
- Studies in Nutrition and Phenotypic Plasticity Unit, Department of Nutrition, Federal University of Pernambuco, Recife, Brazil
| | - Márcia Maria da Silva
- Graduate Program in Nutrition (Posnutri), Health Sciences Center, Federal University of Pernambuco, Recife, Brazil
- Studies in Nutrition and Phenotypic Plasticity Unit, Department of Nutrition, Federal University of Pernambuco, Recife, Brazil
| | - Bárbara J R Costa-de-Santana
- Studies in Nutrition and Phenotypic Plasticity Unit, Department of Nutrition, Federal University of Pernambuco, Recife, Brazil
- Graduate Program in Neuropsychiatry and Behavioral Sciences (Posneuro), Federal University of Pernambuco, Recife, Brazil
| | - Joaci Pereira Dos Santos Junior
- Studies in Nutrition and Phenotypic Plasticity Unit, Department of Nutrition, Federal University of Pernambuco, Recife, Brazil
| | - Luís Miguel Saavedra
- Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, Mexico
| | | | - Juan José Valdéz-Alarcón
- Centro Multidisciplinario de Estudios en Biotecnología - Facultad de Medicina Veterinaria y Zootecnia, Universidad Michoacana de San Nicolás de Hidalgo, Tarímbaro, Mexico
| | - Claudia Jacques Lagranha
- Graduate Program in Biochemistry and Physiology (PGBqF), Federal University of Pernambuco, Recife, Brazil
- Graduate Program in Neuropsychiatry and Behavioral Sciences (Posneuro), Federal University of Pernambuco, Recife, Brazil
| | - Omar Guzman-Quevedo
- Instituto Tecnológico Superior de Tacámbaro, Tacámbaro, Mexico
- Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, Mexico
- Graduate Program in Neuropsychiatry and Behavioral Sciences (Posneuro), Federal University of Pernambuco, Recife, Brazil
| | - Luz Torner
- Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, Mexico
| | - Ana Elisa Toscano
- Graduate Program in Nutrition (Posnutri), Health Sciences Center, Federal University of Pernambuco, Recife, Brazil
- Studies in Nutrition and Phenotypic Plasticity Unit, Department of Nutrition, Federal University of Pernambuco, Recife, Brazil
- Graduate Program in Neuropsychiatry and Behavioral Sciences (Posneuro), Federal University of Pernambuco, Recife, Brazil
- Nursing Unit, Vitória Academic Center, Federal University of Pernambuco, Vitória de Santo Antão, Brazil
| |
Collapse
|
13
|
Lu W, Wen J. H 2S-RhoA/ROCK Pathway and Glial Cells in Axonal Remyelination After Ischemic Stroke. Mol Neurobiol 2023; 60:5493-5504. [PMID: 37322287 DOI: 10.1007/s12035-023-03422-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 06/03/2023] [Indexed: 06/17/2023]
Abstract
Ischemic stroke is one of the main reasons of disability and death. Stroke-induced functional deficits are mainly due to the secondary degeneration of the white matter characterized by axonal demyelination and injury of axon-glial integrity. Enhancement of the axonal regeneration and remyelination could promote the neural functional recovery. However, cerebral ischemia-induced activation of RhoA/Rho kinase (ROCK) pathway plays a crucial and harmful role in the process of axonal recovery and regeneration. Inhibition of this pathway could promote the axonal regeneration and remyelination. In addition, hydrogen sulfide (H2S) has the significant neuroprotective role during the recovery of ischemic stroke via inhibiting the inflammatory response and oxidative stress, regulating astrocyte function, promoting the differentiation of endogenous oligodendrocyte precursor cells (OPCs) to mature oligodendrocyte. Among all of these effects, promoting the formation of mature oligodendrocyte is a crucial part of axonal regeneration and remyelination. Furthermore, numerous studies have uncovered the crosstalk between astrocytes and oligodendrocyte, microglial cells and oligodendrocyte in the axonal remyelination following ischemic stroke. The purpose of this review was to discuss the relationship among H2S, RhoA/ROCK pathway, astrocytes, and microglial cells in the axonal remyelination following ischemic stroke to reveal new strategies for preventing and treating this devastating disease.
Collapse
Affiliation(s)
- Weizhuo Lu
- Medical Branch, Hefei Technology College, Hefei, China
| | - Jiyue Wen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.
| |
Collapse
|
14
|
Lu W, Chen Z, Wen J. The role of RhoA/ROCK pathway in the ischemic stroke-induced neuroinflammation. Biomed Pharmacother 2023; 165:115141. [PMID: 37437375 DOI: 10.1016/j.biopha.2023.115141] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 07/03/2023] [Accepted: 07/07/2023] [Indexed: 07/14/2023] Open
Abstract
It is widely known that ischemic stroke is the prominent cause of death and disability. To date, neuroinflammation following ischemic stroke represents a complex event, which is an essential process and affects the prognosis of both experimental stroke animals and stroke patients. Intense neuroinflammation occurring during the acute phase of stroke contributes to neuronal injury, BBB breakdown, and worse neurological outcomes. Inhibition of neuroinflammation may be a promising target in the development of new therapeutic strategies. RhoA is a small GTPase protein that activates a downstream effector, ROCK. The up-regulation of RhoA/ROCK pathway possesses important roles in promoting the neuroinflammation and mediating brain injury. In addition, nuclear factor-kappa B (NF-κB) is another vital regulator of ischemic stroke-induced neuroinflammation through regulating the functions of microglial cells and astrocytes. After stroke onset, the microglial cells and astrocytes are activated and undergo the morphological and functional changes, thereby deeply participate in a complicated neuroinflammation cascade. In this review, we focused on the relationship among RhoA/ROCK pathway, NF-κB and glial cells in the neuroinflammation following ischemic stroke to reveal new strategies for preventing the intense neuroinflammation.
Collapse
Affiliation(s)
- Weizhuo Lu
- Medical Branch, Hefei Technology College, Hefei, China
| | - Zhiwu Chen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.
| | - Jiyue Wen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.
| |
Collapse
|
15
|
Tan R, Hong R, Sui C, Yang D, Tian H, Zhu T, Yang Y. The role and potential therapeutic targets of astrocytes in central nervous system demyelinating diseases. Front Cell Neurosci 2023; 17:1233762. [PMID: 37720543 PMCID: PMC10502347 DOI: 10.3389/fncel.2023.1233762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 08/21/2023] [Indexed: 09/19/2023] Open
Abstract
Astrocytes play vital roles in the central nervous system, contributing significantly to both its normal functioning and pathological conditions. While their involvement in various diseases is increasingly recognized, their exact role in demyelinating lesions remains uncertain. Astrocytes have the potential to influence demyelination positively or negatively. They can produce and release inflammatory molecules that modulate the activation and movement of other immune cells. Moreover, they can aid in the clearance of myelin debris through phagocytosis and facilitate the recruitment and differentiation of oligodendrocyte precursor cells, thereby promoting axonal remyelination. However, excessive or prolonged astrocyte phagocytosis can exacerbate demyelination and lead to neurological impairments. This review provides an overview of the involvement of astrocytes in various demyelinating diseases, emphasizing the underlying mechanisms that contribute to demyelination. Additionally, we discuss the interactions between oligodendrocytes, oligodendrocyte precursor cells and astrocytes as therapeutic options to support myelin regeneration. Furthermore, we explore the role of astrocytes in repairing synaptic dysfunction, which is also a crucial pathological process in these disorders.
Collapse
Affiliation(s)
- Rui Tan
- Department of Neurosurgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Rui Hong
- Department of Neurosurgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chunxiao Sui
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Tianjin's Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Dianxu Yang
- Department of Neurosurgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hengli Tian
- Department of Neurosurgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tao Zhu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Yang Yang
- Department of Neurosurgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
16
|
Jiang G, Ayaki T, Maki T, Yasuda K, Yoshii D, Kaji S, Takahashi R. Evaluation of BCAS1-positive immature oligodendrocytes after cerebral ischemic stroke and SVD. Neurosci Lett 2023; 812:137405. [PMID: 37479175 DOI: 10.1016/j.neulet.2023.137405] [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: 05/02/2023] [Revised: 07/02/2023] [Accepted: 07/18/2023] [Indexed: 07/23/2023]
Abstract
Ischemic cerebrovascular disease is an important cause of physical disability and dementia. Oligodendrocytes (OLGs), which differentiate from oligodendrocyte precursor cells (OPCs), are crucial for remyelination of the damaged brain and functional recovery. Breast carcinoma amplified sequence 1 (BCAS1) has recently been shown to be highly expressed in newly formed pre-myelinating oligodendrocytes (pre-mOLGs), while its expression level is reduced in mature OLGs. In this study, we analyzed BCAS1 expression by immunohistochemical analysis of human post-mortem brain tissue from six stroke patients (death within 2 months after stroke onset) and eight small vessel disease (SVD) patients. Control post-mortem brain tissue was from eight age-matched patients without any obvious central nervous system (CNS) pathology. The Olig2 expression in the area corresponding to the same section of the BCAS1-stained slice was analyzed to determine the total oligodendrocyte lineage. The percentage of differentiating OPCs in the oligodendrocyte lineage was calculated as the ratio of BCAS1+ to Olig2+ cells (BCAS1+/Olig2+). The stroke and SVD cases showed demyelination with decreased expression of myelin basic protein (MBP, a mature OLG marker). The stroke cases showed significantly increased numbers of early-stage BCAS1+ cells with an immature morphology and Olig2+ cells (pan-oligodendrocyte lineages) in the peri-infarct areas in both the cortex and white matter, but showed no increase in the number of late-stage BCAS1+ cells with a mature morphology. In contrast, the SVD cases showed no significant increase in Olig2+ and BCAS1+ cells. These results indicated that remyelination dysfunction could be attributed to insufficient maturation of OPCs in stroke and impaired recruitment of OPCs in SVD.
Collapse
Affiliation(s)
- Guanhua Jiang
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takashi Ayaki
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Takakuni Maki
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ken Yasuda
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Daisuke Yoshii
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Seiji Kaji
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ryosuke Takahashi
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
17
|
Mihailova V, Stoyanova II, Tonchev AB. Glial Populations in the Human Brain Following Ischemic Injury. Biomedicines 2023; 11:2332. [PMID: 37760773 PMCID: PMC10525766 DOI: 10.3390/biomedicines11092332] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/14/2023] [Accepted: 08/19/2023] [Indexed: 09/29/2023] Open
Abstract
There is a growing interest in glial cells in the central nervous system due to their important role in maintaining brain homeostasis under physiological conditions and after injury. A significant amount of evidence has been accumulated regarding their capacity to exert either pro-inflammatory or anti-inflammatory effects under different pathological conditions. In combination with their proliferative potential, they contribute not only to the limitation of brain damage and tissue remodeling but also to neuronal repair and synaptic recovery. Moreover, reactive glial cells can modulate the processes of neurogenesis, neuronal differentiation, and migration of neurons in the existing neural circuits in the adult brain. By discovering precise signals within specific niches, the regulation of sequential processes in adult neurogenesis holds the potential to unlock strategies that can stimulate the generation of functional neurons, whether in response to injury or as a means of addressing degenerative neurological conditions. Cerebral ischemic stroke, a condition falling within the realm of acute vascular disorders affecting the circulation in the brain, stands as a prominent global cause of disability and mortality. Extensive investigations into glial plasticity and their intricate interactions with other cells in the central nervous system have predominantly relied on studies conducted on experimental animals, including rodents and primates. However, valuable insights have also been gleaned from in vivo studies involving poststroke patients, utilizing highly specialized imaging techniques. Following the attempts to map brain cells, the role of various transcription factors in modulating gene expression in response to cerebral ischemia is gaining increasing popularity. Although the results obtained thus far remain incomplete and occasionally ambiguous, they serve as a solid foundation for the development of strategies aimed at influencing the recovery process after ischemic brain injury.
Collapse
Affiliation(s)
- Victoria Mihailova
- Department of Anatomy and Cell Biology, Faculty of Medicine, Medical University Varna, 9000 Varna, Bulgaria; (I.I.S.); (A.B.T.)
| | | | | |
Collapse
|
18
|
Qiao C, Liu Z, Qie S. The Implications of Microglial Regulation in Neuroplasticity-Dependent Stroke Recovery. Biomolecules 2023; 13:biom13030571. [PMID: 36979506 PMCID: PMC10046452 DOI: 10.3390/biom13030571] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/23/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
Stroke causes varying degrees of neurological deficits, leading to corresponding dysfunctions. There are different therapeutic principles for each stage of pathological development. Neuroprotection is the main treatment in the acute phase, and functional recovery becomes primary in the subacute and chronic phases. Neuroplasticity is considered the basis of functional restoration and neurological rehabilitation after stroke, including the remodeling of dendrites and dendritic spines, axonal sprouting, myelin regeneration, synapse shaping, and neurogenesis. Spatiotemporal development affects the spontaneous rewiring of neural circuits and brain networks. Microglia are resident immune cells in the brain that contribute to homeostasis under physiological conditions. Microglia are activated immediately after stroke, and phenotypic polarization changes and phagocytic function are crucial for regulating focal and global brain inflammation and neurological recovery. We have previously shown that the development of neuroplasticity is spatiotemporally consistent with microglial activation, suggesting that microglia may have a profound impact on neuroplasticity after stroke and may be a key therapeutic target for post-stroke rehabilitation. In this review, we explore the impact of neuroplasticity on post-stroke restoration as well as the functions and mechanisms of microglial activation, polarization, and phagocytosis. This is followed by a summary of microglia-targeted rehabilitative interventions that influence neuroplasticity and promote stroke recovery.
Collapse
Affiliation(s)
- Chenye Qiao
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing 100144, China
| | - Zongjian Liu
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing 100144, China
| | - Shuyan Qie
- Department of Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing 100144, China
| |
Collapse
|
19
|
Koo DL, Cabeen RP, Yook SH, Cen SY, Joo EY, Kim H. More extensive white matter disruptions present in untreated obstructive sleep apnea than we thought: A large sample diffusion imaging study. Hum Brain Mapp 2023; 44:3045-3056. [PMID: 36896706 PMCID: PMC10171547 DOI: 10.1002/hbm.26261] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 12/22/2022] [Accepted: 02/21/2023] [Indexed: 03/11/2023] Open
Abstract
Obstructive sleep apnea (OSA) may lead to white mater (WM) disruptions and cognitive deficits. However, no studies have investigated the full extent of the brain WM, and its associations with cognitive deficits in OSA remain unclear. We thus applied diffusion tensor imaging (DTI) tractography with multi-fiber models and used atlas-based bundle-specific approach to investigate the WM abnormalities for various tracts of the cerebral cortex, thalamus, brainstem, and cerebellum in patients with untreated OSA. We enrolled 100 OSA patients and 63 healthy controls. Fractional anisotropy (FA) and mean diffusivity (MD) values mapped on 33 regions of interest including WM tracts of cortex, thalamus, brainstem, and cerebellum were obtained from tractography-based reconstructions. We compared FA/MD values between groups and correlated FA/MD with clinical data in the OSA group after controlling for age and body mass index. OSA patients showed significantly lower FA values in multiple WM fibers including corpus callosum, inferior fronto-occipital fasciculus, middle/superior longitudinal fasciculi, thalamic radiations, and uncinate (FDR <0.05). Higher FA values were found in medial lemniscus of patients compared to controls (FDR <0.05). Lower FA values of rostrum of corpus callosum correlated with lower visual memory performance in OSA group (p < .005). Our quantitative DTI analysis demonstrated that untreated OSA could negatively impact the integrity of pathways more broadly, including brainstem structures such as medial lemniscus, in comparison to previous findings. Fiber tract abnormalities of the rostral corpus callosum were associated with impaired visual memory in untreated OSA may provide insights into the related pathomechanism.
Collapse
Affiliation(s)
- Dae Lim Koo
- Department of Neurology, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, South Korea
| | - Ryan P Cabeen
- Department of Neurology, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Soon Hyun Yook
- Department of Neurology, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Steven Yong Cen
- Department of Radiology, USC Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Eun Yeon Joo
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute, Seoul, South Korea
| | - Hosung Kim
- Department of Neurology, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
20
|
Kalinichenko SG, Pushchin II, Matveeva NY. Neurotoxic and cytoprotective mechanisms in the ischemic neocortex. J Chem Neuroanat 2023; 128:102230. [PMID: 36603664 DOI: 10.1016/j.jchemneu.2022.102230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/30/2022] [Accepted: 12/30/2022] [Indexed: 01/03/2023]
Abstract
Neuronal damage in ischemic stroke occurs due to permanent imbalance between the metabolic needs of the brain and the ability of the blood-vascular system to maintain glucose delivery and adequate gas exchange. Oxidative stress and excitotoxicity trigger complex processes of neuroinflammation, necrosis, and apoptosis of both neurons and glial cells. This review summarizes data on the structural and chemical changes in the neocortex and main cytoprotective effects induced by focal ischemic stroke. We focus on the expression of neurotrophins (NT) and molecular and cellular changes in neurovascular units in ischemic brain. We also discuss how these factors affect the apoptosis of cortical cells. Ischemic damage involves close interaction of a wide range of signaling molecules, each acting as an efficient marker of cell state in both the ischemic core and penumbra. NTs play the main regulatory role in brain tissue recovery after ischemic injury. Heterogeneous distribution of the BDNF, NT-3, and GDNF immunoreactivity is concordant with the selective response of different types of cortical neurons and glia to ischemic injury and allows mapping the position of viable neurons. Astrocytes are the central link in neurovascular coupling in ischemic brain by providing other cells with a wide range of vasotropic factors. The NT expression coincides with the distribution of reactive astrocytes, marking the boundaries of the penumbra. The development of ischemic stroke is accompanied by a dramatic change in the distribution of GDNF reactivity. In early ischemic period, it is mainly observed in cortical neurons, while in late one, the bulk of GDNF-positive cells are various types of glia, in particular, astrocytes. The proportion of GDNF-positive astrocytes increases gradually throughout the ischemic period. Some factors that exert cytoprotective effects in early ischemic period may display neurotoxic and pro-apoptotic effects later on. The number of apoptotic cells in the ischemic brain tissue correlates with the BDNF levels, corroborating its protective effects. Cytoprotection and neuroplasticity are two lines of brain protection and recovery after ischemic stroke. NTs can be considered an important link in these processes. To develop efficient pharmacological therapy for ischemic brain injury, we have to deepen our understanding of neurochemical adaptation of brain tissue to acute stroke.
Collapse
Affiliation(s)
- Sergei G Kalinichenko
- Department of Histology, Cytology, and Embryology, Pacific State Medical University, Vladivostok 690950, Russia
| | - Igor I Pushchin
- Laboratory of Physiology, A.V. Zhirmusky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok 690041, Russia.
| | - Natalya Yu Matveeva
- Department of Histology, Cytology, and Embryology, Pacific State Medical University, Vladivostok 690950, Russia
| |
Collapse
|
21
|
Effects of HF-rTMS on microglial polarization and white matter integrity in rats with poststroke cognitive impairment. Behav Brain Res 2023; 439:114242. [PMID: 36455674 DOI: 10.1016/j.bbr.2022.114242] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 11/24/2022] [Accepted: 11/27/2022] [Indexed: 11/30/2022]
Abstract
Poststroke cognitive impairment (PSCI) occurs frequently after stroke, but effective treatments are lacking. Previous studies have revealed that high-frequency repetitive transcranial magnetic stimulation (HF-rTMS) has a beneficial effect on PSCI, but the mechanism is unclear. This study aimed to evaluate the effect of 10 and 20 Hz HF-rTMS on PSCI and the possible mechanisms. An ischemic stroke rat model was established by transient middle cerebral artery occlusion (tMCAO). The modified neurological deficit score (mNSS) and Morris water maze tests were conducted to assess neurological function and cognitive function. Luxol Fast Blue (LFB) staining was performed to evaluate white matter damage. Proinflammatory and anti-inflammatory cytokines were measured using enzyme-linked immunosorbent assays (ELISA). Immunofluorescence was used to assess microglial activation and polarization. Western blotting was performed to measure JAK2-STAT3 pathway-related protein expression. We found that HF-rTMS decreased the neurological deficit score. Compared with 10 Hz HF-rTMS, 20 Hz HF-rTMS more markedly improved the cognitive function of tMCAO rats at day 28 after operation. Furthermore, 20 Hz HF-rTMS attenuates white matter lesion, decreased proinflammatory cytokine levels, and increased anti-inflammatory cytokine levels. It also decreased the number of CD68- and CD16/32-positive microglia and increased the number of CD206-positive microglia. In addition, p-JAK2, JAK2, p-STAT3 and STAT3 expression was increased. These findings suggest that HF-rTMS improves cognitive function and attenuates white matter lesion in tMCAO rats by shifting microglia toward the M2 phenotype. Mechanisms may be related to regulation JAK2-STAT3 pathways.
Collapse
|
22
|
Maglinger B, Frank JA, Fraser JF, Pennypacker KR. Reverse Translation to Develop Post-stroke Therapeutic Interventions during Mechanical Thrombectomy: Lessons from the BACTRAC Trial. Methods Mol Biol 2023; 2616:391-402. [PMID: 36715948 DOI: 10.1007/978-1-0716-2926-0_27] [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] [Indexed: 01/31/2023]
Abstract
The majority of strokes, approximately 87%, are ischemic in etiology with the remaining hemorrhagic in origin. Emergent large vessel occlusions (ELVOs) are a subtype of ischemic stroke accounting for approximately 30-40% of acute large vessel blockages. Treatment for ELVOs focuses on recanalization of the occluded vessel by time-sensitive administration of tissue plasminogen activator (tPA) or thrombus removal using mechanical thrombectomy. Although a great deal of time and resources have focused on translational stroke research, little progress has been made in the area of identifying additional new treatments for stroke. Translational limitations include difficulty simulating human comorbid conditions in animal models, as well as the temporal nature of stroke pathology. The Blood And Clot Thrombectomy Registry And Collaboration represents an ongoing tissue registry for thrombectomy patients and includes collection of intracranial arterial blood, systemic arterial blood, thrombi, as well as a series of clinical and radiographic data points for analysis. This chapter will explore the methodologies employed and results obtained from studying BACTRAC-derived human biological specimens and how they can inform translational experimental design in animal studies.
Collapse
Affiliation(s)
- Benton Maglinger
- Department of Neurology, Department of Neuroscience, The University of Kentucky, Lexington, KY, USA
| | - Jacqueline A Frank
- Department of Neurology, Department of Neuroscience, The University of Kentucky, Lexington, KY, USA
- Center for Advanced Translational Stroke Science, University of Kentucky, Lexington, KY, USA
| | - Justin F Fraser
- Department of Neurology, Department of Neuroscience, The University of Kentucky, Lexington, KY, USA
- Center for Advanced Translational Stroke Science, University of Kentucky, Lexington, KY, USA
- Department of Neurosurgery, University of Kentucky, Lexington, KY, USA
- Department of Radiology, University of Kentucky, Lexington, KY, USA
- Department of Neuroscience, University of Kentucky, Lexington, KY, USA
| | - Keith R Pennypacker
- Department of Neurology, Department of Neuroscience, The University of Kentucky, Lexington, KY, USA.
- Center for Advanced Translational Stroke Science, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
23
|
Hey G, Bhutani S, Woolridge M, Patel A, Walls A, Lucke-Wold B. Immunologic Implications for Stroke Recovery: Unveiling the Role of the Immune System in Pathogenesis, Neurorepair, and Rehabilitation. JOURNAL OF CELLULAR IMMUNOLOGY 2023; 5:65-81. [PMID: 37854481 PMCID: PMC10583807 DOI: 10.33696/immunology.5.170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/20/2023]
Abstract
Stroke is a debilitating neurologic condition characterized by an interruption or complete blockage of blood flow to certain areas of the brain. While the primary injury occurs at the time of the initial ischemic event or hemorrhage, secondary injury mechanisms contribute to neuroinflammation, disruption of the blood-brain barrier (BBB), excitotoxicity, and cerebral edema in the days and hours after stroke. Of these secondary mechanisms of injury, significant dysregulation of various immune populations within the body plays a crucial role in exacerbating brain damage after stroke. Pathological activity of glial cells, infiltrating leukocytes, and the adaptive immune system promote neuroinflammation, BBB damage, and neuronal death. Chronic immune activation can additionally encourage the development of neurologic deficits, immunosuppression, and dysregulation of the gut microbiome. As such, immunotherapy has emerged as a promising strategy for the clinical management of stroke in a highly patient-specific manner. These strategies include regulatory T cells (Tregs), cell adhesion molecules, cytokines, and monoclonal antibodies. However, the use of immunotherapy for stroke remains largely in the early stages, highlighting the need for continued research efforts before widespread clinical use.
Collapse
Affiliation(s)
- Grace Hey
- College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Siya Bhutani
- College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Maxwell Woolridge
- College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Aashay Patel
- College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Anna Walls
- College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Brandon Lucke-Wold
- Department of Neurosurgery, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
24
|
Liu Y, Fan L, Jiang X, Lu Y, Li Y. A case study of repetitive transcranial magnetic stimulation for cryptococcal meningitis combined with cognitive impairment. Front Hum Neurosci 2022; 16:1061916. [PMID: 36590060 PMCID: PMC9800931 DOI: 10.3389/fnhum.2022.1061916] [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: 10/05/2022] [Accepted: 11/23/2022] [Indexed: 12/23/2022] Open
Abstract
Cryptococcal meningitis (CM) is a central nervous system disease caused by a novel Cryptococcus infection that leads to subacute or chronic inflammatory changes in the nervous system. In this study, we present the case of a woman aged 72 years with CM and severe cognitive impairment and disabilities. The cognitive assessment indicated that, although her cognitive function was impaired, especially executive function, it largely improved after receiving anti-infectious and repetitive transcranial magnetic stimulation, which can alter the membrane potential of the cortical nerve cells by triggering long-term potentiation and depression, modulating and releasing hormones, reducing the level of neuroinflammatory and peripheral blood cytokines, promoting nerve regeneration and synaptic remodeling, and changing the activity of the neural circuitry of the dorsolateral prefrontal cortex. We argue that this case provides a novel method of treatment for patients with CM in conjunction with cognitive impairments.
Collapse
Affiliation(s)
- Yuanbiao Liu
- Department of Rehabilitation Medicine, Second Affiliated Hospital of Nanjing Medical University, Nanjing, China,*Correspondence: Yuanbiao Liu
| | - Lei Fan
- Department of Rehabilitation Medicine, Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xinlin Jiang
- Department of Rehabilitation Medicine, Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yi Lu
- Physical Medicine and Rehabilitation Unit, Nanjing Medical University, Nanjing, China
| | - Yige Li
- Physical Medicine and Rehabilitation Unit, Nanjing Medical University, Nanjing, China
| |
Collapse
|
25
|
Liu L, Liu J, Li M, Lyu J, Su W, Feng S, Ji X. Selective brain hypothermia attenuates focal cerebral ischemic injury and improves long-term neurological outcome in aged female mice. CNS Neurosci Ther 2022; 29:129-139. [PMID: 36341958 PMCID: PMC9804044 DOI: 10.1111/cns.14017] [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: 06/24/2022] [Revised: 10/09/2022] [Accepted: 10/20/2022] [Indexed: 11/09/2022] Open
Abstract
AIMS This study aimed to investigate the effects of mild selective brain hypothermia on aged female ischemic mice. METHODS A distal middle cerebral artery occlusion (dMCAO) model was established in aged female mice, who were then subjected to mild selective brain hypothermia immediately after the dMCAO procedure. Neurological behavioral examinations were conducted prior to and up to 35 days post-ischemia. Infarct volume, brain atrophy, pro-inflammation, and anti-inflammation microglia/macrophages phenotype and white matter injury were evaluated by immunofluorescence staining. Correlations between neurological behaviors and histological parameters were evaluated by Pearson product linear regression analysis. RESULTS Sensorimotor and cognitive function tests confirmed the protective effect of mild selective brain hypothermia in elderly female ischemic mice. In addition, hypothermia decreased the infarct volume and brain atrophy induced by focal cerebral ischemia. Furthermore, hypothermia alleviated ischemia-induced short-term and long-term white matter injury, which was correlated with behavioral deficits. Finally, hypothermia suppressed the harmful immunological response by promoting the transformation of pro-inflammatory microglia/macrophages to anti-inflammatory phenotype. This polarization was negatively correlated with neuronal loss and white matter injury. CONCLUSION Mild selective brain hypothermia promoted long-term functional recovery by alleviating white matter damage in an aged female mouse model of ischemia.
Collapse
Affiliation(s)
- Liqiang Liu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain DisordersCapital Medical UniversityBeijingChina
| | - Jia Liu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain DisordersCapital Medical UniversityBeijingChina
| | - Ming Li
- Beijing Institute of Geriatrics, Xuanwu HospitalCapital Medical UniversityBeijingChina
| | - Junxuan Lyu
- Department of Neurology, Xuanwu HospitalCapital Medical UniversityBeijingChina
| | - Wei Su
- Department of Neurosurgery, Beijing Tsing Hua Chang Gung Hospital, School of Clinical MedicineTsing Hua UniversityBeijingChina
| | - Shejun Feng
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain DisordersCapital Medical UniversityBeijingChina
| | - Xunming Ji
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain DisordersCapital Medical UniversityBeijingChina,Department of Neurosurgery, Xuanwu HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
26
|
Dietz RM, Dingman AL, Herson PS. Cerebral ischemia in the developing brain. J Cereb Blood Flow Metab 2022; 42:1777-1796. [PMID: 35765984 PMCID: PMC9536116 DOI: 10.1177/0271678x221111600] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 04/29/2022] [Accepted: 05/27/2022] [Indexed: 11/16/2022]
Abstract
Brain ischemia affects all ages, from neonates to the elderly population, and is a leading cause of mortality and morbidity. Multiple preclinical rodent models involving different ages have been developed to investigate the effect of ischemia during different times of key brain maturation events. Traditional models of developmental brain ischemia have focused on rodents at postnatal day 7-10, though emerging models in juvenile rodents (postnatal days 17-25) indicate that there may be fundamental differences in neuronal injury and functional outcomes following focal or global cerebral ischemia at different developmental ages, as well as in adults. Here, we consider the timing of injury in terms of excitation/inhibition balance, oxidative stress, inflammatory responses, blood brain barrier integrity, and white matter injury. Finally, we review translational strategies to improve function after ischemic brain injury, including new ideas regarding neurorestoration, or neural repair strategies that restore plasticity, at delayed time points after ischemia.
Collapse
Affiliation(s)
- Robert M Dietz
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
- Department of Anesthesiology, University of Colorado School of Medicine, Aurora, CO, USA
- Neuronal Injury Program, University of Colorado School of Medicine, Aurora, CO, USA
| | - Andra L Dingman
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
- Neuronal Injury Program, University of Colorado School of Medicine, Aurora, CO, USA
| | - Paco S Herson
- Department of Neurological Surgery, The Ohio State University College of Medicine, Columbus, OH, USA
| |
Collapse
|
27
|
Raffaele S, Fumagalli M. Dynamics of Microglia Activation in the Ischemic Brain: Implications for Myelin Repair and Functional Recovery. Front Cell Neurosci 2022; 16:950819. [PMID: 35899017 PMCID: PMC9309466 DOI: 10.3389/fncel.2022.950819] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
Ischemic stroke is a neurological disorder representing a leading cause of death and permanent disability world-wide, for which effective regenerative treatments are missing. Oligodendrocyte degeneration and consequent myelin disruption are considered major contributing factors to stroke-associated neurological deficits. Therefore, fostering myelin reconstruction by oligodendrocyte precursor cells (OPCs) has emerged as a promising therapeutic approach to enhance functional recovery in stroke patients. A pivotal role in regulating remyelination is played by microglia, the resident immune cells of the brain. Early after stroke, microglial cells exert beneficial functions, promoting OPC recruitment toward the ischemic lesion and preserving myelin integrity. However, the protective features of microglia are lost during disease progression, contributing to remyelination failure. Unveiling the mechanisms driving the pro-remyelination properties of microglia may provide important opportunities for both reducing myelin damage and promoting its regeneration. Here, we summarize recent evidence describing microglia activation kinetics in experimental models of ischemic injury, focusing on the contribution of these innate immune cells to myelin damage and repair. Some molecular signals regulating the pro-regenerative functions of microglia after stroke have been highlighted to provide new possible therapeutic targets involved in the protective functions of these cells. Finally, we analyzed the impact of microglia-to-OPCs communication via extracellular vesicles on post-stroke remyelination and functional recovery. The results collected in this review underline the importance of supporting the pro-remyelination functions of microglial cells after stroke.
Collapse
|
28
|
Peng K, Koduri S, Ye F, Yang J, Keep RF, Xi G, Hua Y. A timeline of oligodendrocyte death and proliferation following experimental subarachnoid hemorrhage. CNS Neurosci Ther 2022; 28:842-850. [PMID: 35150055 PMCID: PMC9062564 DOI: 10.1111/cns.13812] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/25/2022] [Accepted: 01/27/2022] [Indexed: 12/15/2022] Open
Abstract
AIMS White matter (WM) injury is a critical factor associated with worse outcomes following subarachnoid hemorrhage (SAH). However, the detailed pathological changes are not completely understood. This study investigates temporal changes in the corpus callosum (CC), including WM edema and oligodendrocyte death after SAH, and the role of lipocalin-2 (LCN2) in those changes. METHODS Subarachnoid hemorrhage was induced in adult wild-type or LCN2 knockout mice via endovascular perforation. Magnetic resonance imaging was performed 4 hours, 1 day, and 8 days after SAH, and T2 hyperintensity changes within the CC were quantified to represent WM edema. Immunofluorescence staining was performed to evaluate oligodendrocyte death and proliferation. RESULTS Subarachnoid hemorrhage induced significant CC T2 hyperintensity at 4 hours and 1 day that diminished significantly by 8 days post-procedure. Comparing changes between the 4 hours and 1 day, each individual mouse had an increase in CC T2 hyperintensity volume. Oligodendrocyte death was observed at 4 hours, 1 day, and 8 days after SAH induction, and there was progressive loss of mature oligodendrocytes, while immature oligodendrocytes/oligodendrocyte precursor cells (OPCs) proliferated back to baseline by Day 8 after SAH. Moreover, LCN2 knockout attenuated WM edema and oligodendrocyte death at 24 hours after SAH. CONCLUSIONS Subarachnoid hemorrhage leads to T2 hyperintensity change within the CC, which indicates WM edema. Oligodendrocyte death was observed in the CC within 1 day of SAH, with a partial recovery by Day 8. SAH-induced WM injury was alleviated in an LCN2 knockout mouse model.
Collapse
Affiliation(s)
- Kang Peng
- Department of NeurosurgeryUniversity of MichiganAnn ArborMichiganUSA,Department of NeurosurgeryXiangya HospitalCentral South UniversityChangshaChina
| | - Sravanthi Koduri
- Department of NeurosurgeryUniversity of MichiganAnn ArborMichiganUSA
| | - Fenghui Ye
- Department of NeurosurgeryUniversity of MichiganAnn ArborMichiganUSA
| | - Jinting Yang
- Department of NeurosurgeryUniversity of MichiganAnn ArborMichiganUSA
| | - Richard F. Keep
- Department of NeurosurgeryUniversity of MichiganAnn ArborMichiganUSA
| | - Guohua Xi
- Department of NeurosurgeryUniversity of MichiganAnn ArborMichiganUSA
| | - Ya Hua
- Department of NeurosurgeryUniversity of MichiganAnn ArborMichiganUSA
| |
Collapse
|
29
|
Guo YS, Yuan M, Han Y, Shen XY, Gao ZK, Bi X. Effects of enriched environment on microglia and functional white matter recovery in rats with post stroke cognitive impairment. Neurochem Int 2022; 154:105295. [PMID: 35121010 DOI: 10.1016/j.neuint.2022.105295] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 12/11/2021] [Accepted: 01/27/2022] [Indexed: 01/02/2023]
Abstract
BACKGROUND White matter damage is an important contributor to cognitive impairment after stroke. This study was designed to explore the beneficial effects of enriched environment (EE) on white matter recovery and cognitive dysfunction after stroke, and further explore the potential mechanism of EE on white matter recovery from the perspective of microglia and microglia-mediated neuroinflammation. METHODS Male SD rats underwent middle cerebral artery occlusion(MCAO) or sham surgery. During the MCAO operation, a laser Doppler blood flow meter was used to monitor the blood flow to ensure the success of the model. At 72 hours after the operation, 3 rats were selected for TTC staining to identify the infarct size. One week after surgery, the rats were randomly assigned into four different groups-MCAO+standard environment (SE), MCAO+enriched environment(EE), Sham+SE and Sham+EE for 4 weeks. At four weeks after MCAO surgery, neurological function deficiency condition and cognitive function were assessed using Longa score and Morris Water Maze prior to euthanasia. The loss or regeneration of myelin was stained with LFB, the expression of myelin regeneration-related protein and microglia protein was quantified by western blot and immunofluorescence, and the level of inflammatory factors was measured by ELISA. RESULTS EE treatment remarkably decreased the neurological deficit score, ameliorated the cognitive functional deficit in MCAO rats. Furthermore, EE alleviated white matter lesions and demyelination, increased myelin basic protein expression and decreased the number of activated microglia in the hippocampus of MCAO rats. In addition, ELISA analysis indicated that EE decreased the level of IL-1β, IL-6, which further suggests that EE may reduce the level of pro-inflammatory factors by affecting the expression of microglia marker, IBA1, provide a benefit physiological environment for myelin recovery, and improve post stroke cognitive impairment. CONCLUSIONS Our results suggest that exposure to EE substantially reduced the damage to brain tissue caused by activation of microglia activation, decreased the level of pro-inflammatory cytokins, which may induced by microglia, protected and promote white matter recovery to improve cognitive function after stroke. Our findings also indicate exposure to EE is beneficial for patients with white matter impairment characterised by white matter disease-related inflammation.
Collapse
Affiliation(s)
- Yi-Sha Guo
- Department of Rehabilitation Medicine, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, 201318, China; Shanghai University of Sport, Shanghai, 200438, China
| | - Mei Yuan
- Department of Rehabilitation Medicine, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, 201318, China; Shanghai University of Sport, Shanghai, 200438, China
| | - Yu Han
- Department of Rehabilitation Medicine, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, 201318, China; Shanghai University of Sport, Shanghai, 200438, China
| | - Xin-Ya Shen
- Department of Rehabilitation Medicine, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, 201318, China; Shanghai University of Traditionary Chinese Medicine, Shanghai, 201203, China
| | - Zhen-Kun Gao
- Department of Rehabilitation Medicine, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, 201318, China; Shanghai University of Traditionary Chinese Medicine, Shanghai, 201203, China
| | - Xia Bi
- Department of Rehabilitation Medicine, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, 201318, China; Shanghai University of Sport, Shanghai, 200438, China.
| |
Collapse
|
30
|
Yoshii D, Ayaki T, Wada T, Ozaki A, Yamamoto T, Miyagi Y, Senzaki H, Takahashi R. An autopsy case of adult‐onset neuronal intranuclear inclusion disease with perivascular preservation in cerebral white matter. Neuropathology 2021; 42:66-73. [DOI: 10.1111/neup.12778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 07/21/2021] [Accepted: 08/24/2021] [Indexed: 11/30/2022]
Affiliation(s)
- Daisuke Yoshii
- Department of Neurology Kyoto University Graduate School of Medicine Kyoto Japan
| | - Takashi Ayaki
- Department of Neurology Kyoto University Graduate School of Medicine Kyoto Japan
| | - Takafumi Wada
- Department of Neurology Osaka Saiseikai Nakatsu Hospital Osaka Japan
| | - Akihiko Ozaki
- Department of Neurology Osaka Saiseikai Nakatsu Hospital Osaka Japan
| | - Toru Yamamoto
- Department of Neurology Osaka Saiseikai Nakatsu Hospital Osaka Japan
| | - Yoshimi Miyagi
- Department of Pathology Osaka Saiseikai Nakatsu Hospital Osaka Japan
| | - Hideto Senzaki
- Department of Pathology Osaka Saiseikai Nakatsu Hospital Osaka Japan
| | - Ryosuke Takahashi
- Department of Neurology Kyoto University Graduate School of Medicine Kyoto Japan
| |
Collapse
|
31
|
Spears RC, McLouth CJ, Pennypacker KR, Frank JA, Maglinger B, Martha S, Trout AL, Roberts J, Stowe AM, Sheikhi L, Pahwa S, Fraser JF. Alterations in Local Peri-Infarct Blood Gases in Stroke Patients Undergoing Thrombectomy. World Neurosurg 2021; 158:e317-e322. [PMID: 34728392 DOI: 10.1016/j.wneu.2021.10.171] [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: 06/14/2021] [Revised: 10/25/2021] [Accepted: 10/26/2021] [Indexed: 11/16/2022]
Abstract
BACKGROUND Ischemic stroke is a prevalent, devastating disease with high morbidity and mortality. Despite extensive research using animal models, significant gaps remain in understanding the pathological processes in human stroke. We previously developed a tissue bank to analyze the blood immediately proximal and distal to an intracranial thrombus in patients undergoing mechanical thrombectomy (ClinicalTrials.gov identifier, NCT03153683). Our goal for the present project was to evaluate the blood gas changes and acid/base balance during stroke and determine how vascular collateralization affects these changes. METHODS We analyzed the blood samples and computed tomography angiography collateral scores from the first 62 patients in the BACTRAC (Blood and Clot Thrombectomy Registry and Collaboration) registry. The bicarbonate, partial pressure of oxygen, and partial pressure of carbon dioxide (pCO2) values of the intracranial (distal) and systemic (proximal) arterial blood relative to the occlusive thrombus were analyzed. Analysis of the group differences in systemic and intracranial blood gas values was also performed. RESULTS The partial pressure of oxygen, pCO2, and bicarbonate levels were all significantly higher in the systemic blood than in the intracranial blood (P < 0.001 for all) at thrombectomy. Collateralization did not significantly affect the distal blood gas values. Compared with the female patients, the male patients had had higher systemic pCO2 values (39.8 vs. 36.6 mm Hg; P = 0.0065) and lower systemic and intracranial pH values (7.351 vs. 7.392; P = 0.0047). CONCLUSIONS The arterial blood gases differed immediately proximal and distal to thrombi in large vessel occlusive stroke. Although vascular collateralization did not appear to affect the blood gas changes, some blood gas values differed between men and women. The changes in bicarbonate and pCO2 suggested a compensatory acid-base process occurring at the time of infarction.
Collapse
Affiliation(s)
| | - Christopher J McLouth
- Department of Behavioral Science, University of Kentucky Hospital, Lexington, Kentucky, USA
| | - Keith R Pennypacker
- Department of Neurology and Neuroscience, University of Kentucky, Lexington, Kentucky, USA
| | - Jacqueline A Frank
- Center for Advanced Translational Stroke Science, University of Kentucky, Lexington, Kentucky, USA
| | - Benton Maglinger
- Department of Neurology, University of Kentucky Hospital, Lexington, Kentucky, USA
| | - Sarah Martha
- College of Nursing, University of Kentucky, Lexington, Kentucky, USA; Present address: Sarah Martha, Department of Biobehavioral Nursing Science, College of Nursing, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Amanda L Trout
- Department of Neurology and Neuroscience, University of Kentucky, Lexington, Kentucky, USA
| | - Jill Roberts
- Department of Neurology and Neuroscience, University of Kentucky, Lexington, Kentucky, USA; Center for Advanced Translational Stroke Science, University of Kentucky, Lexington, Kentucky, USA
| | - Ann M Stowe
- Department of Neurology and Neuroscience, University of Kentucky, Lexington, Kentucky, USA; Center for Advanced Translational Stroke Science, University of Kentucky, Lexington, Kentucky, USA
| | - Lila Sheikhi
- Department of Neurosurgery, University of Kentucky, Lexington, Kentucky, USA; Department of Neurology, University of Kentucky, Lexington, Kentucky, USA; Department of Radiology, University of Kentucky, Lexington, Kentucky, USA
| | - Shivani Pahwa
- Department of Neurosurgery, University of Kentucky, Lexington, Kentucky, USA; Department of Neurology, University of Kentucky, Lexington, Kentucky, USA; Department of Radiology, University of Kentucky, Lexington, Kentucky, USA
| | - Justin F Fraser
- Center for Advanced Translational Stroke Science, University of Kentucky, Lexington, Kentucky, USA; Department of Neurosurgery, University of Kentucky, Lexington, Kentucky, USA; Department of Neurology, University of Kentucky, Lexington, Kentucky, USA; Department of Radiology, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
32
|
Sun K, Zhang J, Yang Q, Zhu J, Zhang X, Wu K, Li Z, Xie W, Luo X. Dexmedetomidine exerts a protective effect on ischemic brain injury by inhibiting the P2X7R/NLRP3/Caspase-1 signaling pathway. Brain Res Bull 2021; 174:11-21. [PMID: 33991606 DOI: 10.1016/j.brainresbull.2021.05.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 04/30/2021] [Accepted: 05/08/2021] [Indexed: 12/29/2022]
Abstract
Dexmedetomidine (Dex) has been suggested to exert a protective function in ischemic brain injury. In this study, we aimed to elucidate the intrinsic mechanisms of Dex in regulating microglia pyroptosis in ischemic brain injury via the purinergic 2X7 receptor (P2X7R)/NLRP3/Caspase-1 signaling pathway. First, permanent middle cerebral artery occlusion (p-MCAO) rat model was established, followed by the measurement of behavioral deficit, neuronal injury, the volume of brain edema and the infarct size. Dex treatment was suggested to alleviate the neurological deficits in p-MCAO rats and reduce the brain water content and infarct size. Additionally, rat microglia were cultured in vitro and a model of oxygen and glucose (OGD) was established. Microglia cell activity and ultrastructure were detected. Dex could increase cell activity and reduce LDH activity, partially reversing the changes in cell morphology. Furthermore, the activation of P2X7R/NLRP3/Caspase-1 pathway was tested. The obtained findings indicated Dex suppressed microglial pyroptosis by inhibiting the P2X7R/NLRP3/Caspase-1 pathway. Inhibition of P2X7R or NLRP3 could inhibit Caspase-1 p10 expression, improve cell activity, and reduce LDH activity. The same result was verified in vivo experiments. This study indicated that Dex inhibited microglia pyroptosis by blocking the P2X7R/NLRP3/Caspase-1 pathway, thus playing a protective role against ischemic brain injury.
Collapse
Affiliation(s)
- Ke Sun
- Department of Neurology, Anyang People's Hospital, Anyang, Henan, 455000, China
| | - Jiangang Zhang
- Department of Neurology, Anyang People's Hospital, Anyang, Henan, 455000, China
| | - Qingcheng Yang
- Department of Neurology, Anyang People's Hospital, Anyang, Henan, 455000, China.
| | - Jinzhao Zhu
- Department of Neurology, Anyang People's Hospital, Anyang, Henan, 455000, China
| | - Xiangdong Zhang
- Department of Neurology, Anyang People's Hospital, Anyang, Henan, 455000, China
| | - Kun Wu
- Department of Neurology, Anyang People's Hospital, Anyang, Henan, 455000, China
| | - Zhenhua Li
- Department of Neurology, Anyang People's Hospital, Anyang, Henan, 455000, China
| | - Weizheng Xie
- Department of Neurology, Anyang People's Hospital, Anyang, Henan, 455000, China
| | - Xue Luo
- Department of Neurology, Anyang People's Hospital, Anyang, Henan, 455000, China
| |
Collapse
|
33
|
Li S, Rao JH, Lan XY, Li X, Chu CY, Liang Y, Janowski M, Zhang HT, Walczak P. White matter demyelination predates axonal injury after ischemic stroke in cynomolgus monkeys. Exp Neurol 2021; 340:113655. [PMID: 33617887 DOI: 10.1016/j.expneurol.2021.113655] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/11/2021] [Accepted: 02/16/2021] [Indexed: 02/02/2023]
Abstract
Unraveling the pathology of stroke is a prerequisite for designing therapeutic strategies. It was reported that myelin injury exceeded axonal loss in the peri-infarct region of rodent white matter stroke. An in-depth investigation of the post-stroke white matter damage in higher-order species might innovate stroke intervention. In this study, adult male cynomolgus monkeys received surgical middle cerebral artery occlusion (MCAO), and serial magnetic resonance scans to non-invasively assess brain damage. Spontaneous movements were recorded to evaluate post-stroke behavior. The axon and myelin loss, as well as immune cell infiltration were examined using immunohistochemistry. Magnetic resonance imaging revealed cerebral infarcts and white matter injury after MCAO in monkeys, which were confirmed by neurological deficits. Immunostaining of white matter fibers showed substantial demyelination whilst retention of axons in the infarcts 8 days post MCAO, while a progressive loss of myelin and axons was observed after one month. Gliosis, microglia activation, and leukocyte infiltration were identified in the lesions. These results demonstrate that demyelination predates axonal injury in non-human primate ischemic stroke, which provides a time window for stroke intervention focusing on prevention of progressive axonal loss through myelin regeneration.
Collapse
Affiliation(s)
- Shen Li
- Department of Neurology, Dalian Municipal Central Hospital affiliated with Dalian Medical University, Dalian, China
| | - Jun-Hua Rao
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangdong, China
| | - Xiao-Yan Lan
- Department of Neurology, Dalian Municipal Central Hospital affiliated with Dalian Medical University, Dalian, China; Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland Baltimore, Baltimore, MD, USA
| | - Xu Li
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Cheng-Yan Chu
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland Baltimore, Baltimore, MD, USA
| | - Yajie Liang
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland Baltimore, Baltimore, MD, USA
| | - Miroslaw Janowski
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland Baltimore, Baltimore, MD, USA
| | - Hong-Tian Zhang
- Bayi Brain Hospital affiliated with the 7th Medical Center of PLA General Hospital, Beijing, China.
| | - Piotr Walczak
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland Baltimore, Baltimore, MD, USA.
| |
Collapse
|
34
|
Cellular and Molecular Mechanisms of R/S-Roscovitine and CDKs Related Inhibition under Both Focal and Global Cerebral Ischemia: A Focus on Neurovascular Unit and Immune Cells. Cells 2021; 10:cells10010104. [PMID: 33429982 PMCID: PMC7827530 DOI: 10.3390/cells10010104] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/29/2020] [Accepted: 01/05/2021] [Indexed: 12/29/2022] Open
Abstract
Ischemic stroke is the second leading cause of death worldwide. Following ischemic stroke, Neurovascular Unit (NVU) inflammation and peripheral leucocytes infiltration are major contributors to the extension of brain lesions. For a long time restricted to neurons, the 10 past years have shown the emergence of an increasing number of studies focusing on the role of Cyclin-Dependent Kinases (CDKs) on the other cells of NVU, as well as on the leucocytes. The most widely used CDKs inhibitor, (R)-roscovitine, and its (S) isomer both decreased brain lesions in models of global and focal cerebral ischemia. We previously showed that (S)-roscovitine acted, at least, by modulating NVU response to ischemia. Interestingly, roscovitine was shown to decrease leucocytes-mediated inflammation in several inflammatory models. Specific inhibition of roscovitine majors target CDK 1, 2, 5, 7, and 9 showed that these CDKs played key roles in inflammatory processes of NVU cells and leucocytes after brain lesions, including ischemic stroke. The data summarized here support the investigation of roscovitine as a potential therapeutic agent for the treatment of ischemic stroke, and provide an overview of CDK 1, 2, 5, 7, and 9 functions in brain cells and leucocytes during cerebral ischemia.
Collapse
|
35
|
Ostroumova TM, Ostroumova OD, Parfenov VA, Perepelova EM, Perepelov VA, Kochetkov AI. Effect of Perindopril/Indapamide on Cerebral Blood Flow in Middle-Aged, Treatment-Naïve Patients with Hypertension. Adv Ther 2020; 37:4930-4943. [PMID: 33026579 DOI: 10.1007/s12325-020-01515-7] [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: 08/14/2020] [Accepted: 09/23/2020] [Indexed: 11/24/2022]
Abstract
INTRODUCTION The relationship between blood pressure (BP) and cerebral blood flow (CBF) is not fully understood. This study evaluated the impact of a perindopril arginine/indapamide (Pa/I) single-pill combination (SPC) on CBF in middle-aged patients. METHODS A total of 22 treatment-naïve patients with essential hypertension and at least one hypertension-mediated organ damage and 41 healthy controls were enrolled. At baseline, all participants underwent brain magnetic resonance imaging (MRI); patients with hypertension underwent an additional MRI at end of follow-up. Arterial spin labeling (ASL) was used to calculate CBF in the frontal lobe cortical plate. Patients with hypertension received once-daily Pa/I 5 mg/1.25 mg SPC, which could be increased to Pa/I 10 mg/2.5 mg at 2 weeks if necessary. Patients with hypertension underwent 24-h ambulatory BP monitoring (ABPM) at baseline and end of follow-up. RESULTS Mean baseline BP values were 146.2/93.1 and 119.1/76.1 mmHg in the hypertension and control groups, respectively. Patients with hypertension had significantly (p < 0.001) lower CBF in the cortical plate of both left (36.2 ± 8.3 vs. 45.3 ± 3.5 ml/100 g/min) and right (37.9 ± 7.9 vs. 45.8 ± 3.2 ml/100 g/min) frontal lobes compared to normotensive controls. At the end of follow-up, there was a statistically significant (p < 0.001) increase in CBF in the cortical plate of both left (from 36.2 ± 8.3 to 47.5 ± 9.8 ml/100 g/min) and right frontal lobes (from 37.9 ± 7.9 to 47.4 ± 10.1 ml/100 g/min) compared to baseline. No significant difference was found between end of follow-up CBF levels in frontal lobes of patients with hypertension and those of healthy controls at baseline. Office BP decreased by 24.2/15.5 mmHg and 24-h ABPM from 145.5/95.3 to 120.8/79.3 mmHg. CONCLUSION In middle-aged, treatment-naïve patients with hypertension, Pa/I SPC was associated with increased CBF in the cortical plate of the frontal lobes, which achieved levels of normotensive controls. The increase in CBF had no clear association with observed BP changes. REGISTRATION NUMBER ISRCTN67799751.
Collapse
Affiliation(s)
- Tatiana M Ostroumova
- Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia.
| | - Olga D Ostroumova
- Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
- Federal State Budgetary Educational Institution of Further Professional Education "Russian Medical Academy of Continuous Professional Education" of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
| | - Vladimir A Parfenov
- Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - Elena M Perepelova
- Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - Vsevolod A Perepelov
- Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - Alexey I Kochetkov
- Federal State Budgetary Educational Institution of Further Professional Education "Russian Medical Academy of Continuous Professional Education" of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
| |
Collapse
|
36
|
Barhwal KK, Biswal S, Chandra Nag T, Chaurasia OP, Hota SK. Class switching of carbonic anhydrase isoforms mediates remyelination in CA3 hippocampal neurons during chronic hypoxia. Free Radic Biol Med 2020; 161:102-114. [PMID: 33035636 DOI: 10.1016/j.freeradbiomed.2020.09.029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 09/19/2020] [Accepted: 09/29/2020] [Indexed: 12/17/2022]
Abstract
Chronic exposure to hypoxia results in cerebral white matter hyperintensities, increased P300 latency, delayed response and impairment in working memory. Despite burgeoning evidence on role of myelination in nerve conduction, the effect of chronic hypoxia on myelination of hippocampal neurons has been less studied. The present study provides novel evidence on alterations in myelination of hippocampal CA3 neurons following chronic hypoxic exposure. Sprague Dawley rats exposed to global hypobaric hypoxia simulating altitude of 25,000 ft showed progressive demyelination in CA3 hippocampal neurons on 14 days followed by remyelination on 21 and 28 days. The demyelination of CA3 neurons was associated with increased apoptosis of both oligodendrocyte precursor cells (OPCs) and mature oligodendrocytes (OLs), peroxidation of myelin lipids, and nitration induced reduced expression of Carbonic Anhydrase II (CAII). Prolonged hypoxic exposure of 21 and 28 days on the other hand resulted in peroxisome proliferator-activated receptor alpha (PPARα) induced upregulation of Carbonic Anhydrase IV (CAIV) expression in mature oligodendrocytes through iNOS mediated mechanisms along with reduction in lipid peroxidation and remyelination. Inhibition of carbonic anhydrase activity on the other hand prevented remyelination of CA3 neurons. Based on these findings we propose a novel iNOS mediated mechanism for regulation of myelination in hypoxic hippocampal neurons through class switching of carbonic anhydrases.
Collapse
Affiliation(s)
- Kalpana Kumari Barhwal
- Department of Physiology, All India Institute of Medical Sciences, Bhubaneswar, Odisha, 751019, India.
| | - Suryanarayan Biswal
- Centre for Brain Development and Repair, Institute of Stem Cell Biology and Regenerative Medicine, Bangalore, 560065, India; Defence Institute of High Altitude Research, DRDO, C/o 56 APO, Leh-Ladakh, Jammu & Kashmir, 901205, India
| | - Tapas Chandra Nag
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Om Prakash Chaurasia
- Defence Institute of High Altitude Research, DRDO, C/o 56 APO, Leh-Ladakh, Jammu & Kashmir, 901205, India
| | - Sunil Kumar Hota
- O/o Director General (Life Sciences), DRDO Head Quarters, Rajaji Marg, New Delhi, 110011, India
| |
Collapse
|
37
|
Vaes JEG, Brandt MJV, Wanders N, Benders MJNL, de Theije CGM, Gressens P, Nijboer CH. The impact of trophic and immunomodulatory factors on oligodendrocyte maturation: Potential treatments for encephalopathy of prematurity. Glia 2020; 69:1311-1340. [PMID: 33595855 PMCID: PMC8246971 DOI: 10.1002/glia.23939] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 11/11/2020] [Accepted: 11/12/2020] [Indexed: 12/11/2022]
Abstract
Encephalopathy of prematurity (EoP) is a major cause of morbidity in preterm neonates, causing neurodevelopmental adversities that can lead to lifelong impairments. Preterm birth-related insults, such as cerebral oxygen fluctuations and perinatal inflammation, are believed to negatively impact brain development, leading to a range of brain abnormalities. Diffuse white matter injury is a major hallmark of EoP and characterized by widespread hypomyelination, the result of disturbances in oligodendrocyte lineage development. At present, there are no treatment options available, despite the enormous burden of EoP on patients, their families, and society. Over the years, research in the field of neonatal brain injury and other white matter pathologies has led to the identification of several promising trophic factors and cytokines that contribute to the survival and maturation of oligodendrocytes, and/or dampening neuroinflammation. In this review, we discuss the current literature on selected factors and their therapeutic potential to combat EoP, covering a wide range of in vitro, preclinical and clinical studies. Furthermore, we offer a future perspective on the translatability of these factors into clinical practice.
Collapse
Affiliation(s)
- Josine E G Vaes
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands.,Department of Neonatology, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands
| | - Myrna J V Brandt
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands
| | - Nikki Wanders
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands
| | - Manon J N L Benders
- Department of Neonatology, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands
| | - Caroline G M de Theije
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands
| | | | - Cora H Nijboer
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
38
|
Qiu H, Qian T, Wu T, Wang X, Zhu C, Chen C, Wang L. Umbilical cord blood cells for the treatment of preterm white matter injury: Potential effects and treatment options. J Neurosci Res 2020; 99:778-792. [PMID: 33207392 DOI: 10.1002/jnr.24751] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 12/31/2022]
Abstract
Preterm birth is a global public health problem. A large number of preterm infants survive with preterm white matter injury (PWMI), which leads to neurological deficits, and has multifaceted etiology, clinical course, monitoring, and outcomes. The principal upstream insults leading to PWMI initiation are hypoxia-ischemia and infection and/or inflammation and the key target cells are late oligodendrocyte precursor cells. Current PWMI treatments are mainly supportive, and thus have little effect in terms of protecting the immature brain or repairing injury to improve long-term outcomes. Umbilical cord blood (UCB) cells comprise abundant immunomodulatory and stem cells, which have the potential to reduce brain injury, mainly due to anti-inflammatory and immunomodulatory mechanisms, and also through their release of neurotrophic or growth factors to promote endogenous neurogenesis. In this review, we briefly summarize PWMI pathogenesis and pathophysiology, and the specific properties of different cell types in UCB. We further explore the potential mechanism by which UCB can be used to treat PWMI, and discuss the advantages of and potential issues related to UCB cell therapy. Finally, we suggest potential future studies of UCB cell therapy in preterm infants.
Collapse
Affiliation(s)
- Han Qiu
- Key Laboratory of Neonatal Diseases of Health Commission of the People's Republic of China, Shanghai, China.,Department of Neonatology, National Children's Medical Center/Children's Hospital of Fudan University, Shanghai, China
| | - Tianyang Qian
- Key Laboratory of Neonatal Diseases of Health Commission of the People's Republic of China, Shanghai, China.,Department of Neonatology, National Children's Medical Center/Children's Hospital of Fudan University, Shanghai, China
| | - Tong Wu
- Key Laboratory of Neonatal Diseases of Health Commission of the People's Republic of China, Shanghai, China.,Department of Neonatology, National Children's Medical Center/Children's Hospital of Fudan University, Shanghai, China
| | - Xiaoyang Wang
- Center of Perinatal Medicine and Health, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Changlian Zhu
- Center of Perinatal Medicine and Health, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Henan Key Laboratory of Child Brain Injury, Institute of Neuroscience and Third Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Chao Chen
- Key Laboratory of Neonatal Diseases of Health Commission of the People's Republic of China, Shanghai, China.,Department of Neonatology, National Children's Medical Center/Children's Hospital of Fudan University, Shanghai, China
| | - Laishuan Wang
- Key Laboratory of Neonatal Diseases of Health Commission of the People's Republic of China, Shanghai, China.,Department of Neonatology, National Children's Medical Center/Children's Hospital of Fudan University, Shanghai, China
| |
Collapse
|
39
|
Livingston JM, Syeda T, Christie T, Gilbert EA, Morshead CM. Subacute metformin treatment reduces inflammation and improves functional outcome following neonatal hypoxia ischemia. Brain Behav Immun Health 2020; 7:100119. [PMID: 34589876 PMCID: PMC8474427 DOI: 10.1016/j.bbih.2020.100119] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/21/2020] [Accepted: 07/23/2020] [Indexed: 02/03/2023] Open
Abstract
Hypoxia-ischemia (HI) injury is a leading cause of neonatal death and long-term disability, and existing treatment options for HI offer only modest benefit. Early intervention with the drug metformin has been shown to promote functional improvement in numerous rodent models of injury and has pleiotropic cellular effects in the brain. We have previously shown that 1 week of metformin treatment initiated 24 h after HI in neonatal mice resulted in improved motor and cognitive performance, activation of endogenous neural precursor cells (NPCs), and increased oligodendrogenesis. While promising, a limitation to this work is that immediate pharmacological intervention is not always possible in the clinic. Herein, we investigated whether delaying metformin treatment to begin in the subacute phase post-HI would still effectively promote recovery. Male and female C57/BL6 mice received HI injury postnatally, and metformin treatment began 7 days post-HI for up to 4 weeks. Motor and cognitive performance was assessed across time using behavioural tests (cylinder, foot fault, puzzle box). We found that metformin improved motor and cognitive behaviour, decreased inflammation, and increased oligodendrocytes in the motor cortex. Our present findings demonstrate that a clinically relevant subacute metformin treatment paradigm affords the potential to treat neonatal HI, and that improved outcomes occur through modulation of the inflammatory response and oligodendrogenesis. Subacute metformin treatment improves functional recovery after neonatal hypoxia ischemia. Metformin reduces the number of microglia present in the brain early after injury. Metformin increases the number of oligodendrocytes present in the chronic post-injury phase. Metformin treatment has therapeutic potential in the treatment of hypoxic ischemic brain damage.
Collapse
Affiliation(s)
- Jessica M. Livingston
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, M5S1A8, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, M5S3E, Canada
| | - Tasfia Syeda
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, M5S3E, Canada
| | - Taryn Christie
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, M5S3E, Canada
| | - Emily A.B. Gilbert
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, M5S1A8, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, M5S3E, Canada
| | - Cindi M. Morshead
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, M5S1A8, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, M5S3E, Canada
- Institute of Medical Science, University of Toronto, Toronto, M5S1A8, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, M5S3E1, Canada
- Corresponding author. Faculty of Medicine, Donnelly Centre, University of Toronto, 160 College Street, Toronto, ON, M5S 3E1, Canada.
| |
Collapse
|
40
|
Belov Kirdajova D, Kriska J, Tureckova J, Anderova M. Ischemia-Triggered Glutamate Excitotoxicity From the Perspective of Glial Cells. Front Cell Neurosci 2020; 14:51. [PMID: 32265656 PMCID: PMC7098326 DOI: 10.3389/fncel.2020.00051] [Citation(s) in RCA: 232] [Impact Index Per Article: 46.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 02/21/2020] [Indexed: 12/21/2022] Open
Abstract
A plethora of neurological disorders shares a final common deadly pathway known as excitotoxicity. Among these disorders, ischemic injury is a prominent cause of death and disability worldwide. Brain ischemia stems from cardiac arrest or stroke, both responsible for insufficient blood supply to the brain parenchyma. Glucose and oxygen deficiency disrupts oxidative phosphorylation, which results in energy depletion and ionic imbalance, followed by cell membrane depolarization, calcium (Ca2+) overload, and extracellular accumulation of excitatory amino acid glutamate. If tight physiological regulation fails to clear the surplus of this neurotransmitter, subsequent prolonged activation of glutamate receptors forms a vicious circle between elevated concentrations of intracellular Ca2+ ions and aberrant glutamate release, aggravating the effect of this ischemic pathway. The activation of downstream Ca2+-dependent enzymes has a catastrophic impact on nervous tissue leading to cell death, accompanied by the formation of free radicals, edema, and inflammation. After decades of “neuron-centric” approaches, recent research has also finally shed some light on the role of glial cells in neurological diseases. It is becoming more and more evident that neurons and glia depend on each other. Neuronal cells, astrocytes, microglia, NG2 glia, and oligodendrocytes all have their roles in what is known as glutamate excitotoxicity. However, who is the main contributor to the ischemic pathway, and who is the unsuspecting victim? In this review article, we summarize the so-far-revealed roles of cells in the central nervous system, with particular attention to glial cells in ischemia-induced glutamate excitotoxicity, its origins, and consequences.
Collapse
Affiliation(s)
- Denisa Belov Kirdajova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic (ASCR), Prague, Czechia.,Second Faculty of Medicine, Charles University, Prague, Czechia
| | - Jan Kriska
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic (ASCR), Prague, Czechia.,Second Faculty of Medicine, Charles University, Prague, Czechia
| | - Jana Tureckova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic (ASCR), Prague, Czechia
| | - Miroslava Anderova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic (ASCR), Prague, Czechia.,Second Faculty of Medicine, Charles University, Prague, Czechia
| |
Collapse
|
41
|
Xu S, Lu J, Shao A, Zhang JH, Zhang J. Glial Cells: Role of the Immune Response in Ischemic Stroke. Front Immunol 2020; 11:294. [PMID: 32174916 PMCID: PMC7055422 DOI: 10.3389/fimmu.2020.00294] [Citation(s) in RCA: 377] [Impact Index Per Article: 75.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 02/05/2020] [Indexed: 12/16/2022] Open
Abstract
Ischemic stroke, which accounts for 75-80% of all strokes, is the predominant cause of morbidity and mortality worldwide. The post-stroke immune response has recently emerged as a new breakthrough target in the treatment strategy for ischemic stroke. Glial cells, including microglia, astrocytes, and oligodendrocytes, are the primary components of the peri-infarct environment in the central nervous system (CNS) and have been implicated in post-stroke immune regulation. However, increasing evidence suggests that glial cells exert beneficial and detrimental effects during ischemic stroke. Microglia, which survey CNS homeostasis and regulate innate immune responses, are rapidly activated after ischemic stroke. Activated microglia release inflammatory cytokines that induce neuronal tissue injury. By contrast, anti-inflammatory cytokines and neurotrophic factors secreted by alternatively activated microglia are beneficial for recovery after ischemic stroke. Astrocyte activation and reactive gliosis in ischemic stroke contribute to limiting brain injury and re-establishing CNS homeostasis. However, glial scarring hinders neuronal reconnection and extension. Neuroinflammation affects the demyelination and remyelination of oligodendrocytes. Myelin-associated antigens released from oligodendrocytes activate peripheral T cells, thereby resulting in the autoimmune response. Oligodendrocyte precursor cells, which can differentiate into oligodendrocytes, follow an ischemic stroke and may result in functional recovery. Herein, we discuss the mechanisms of post-stroke immune regulation mediated by glial cells and the interaction between glial cells and neurons. In addition, we describe the potential roles of various glial cells at different stages of ischemic stroke and discuss future intervention targets.
Collapse
Affiliation(s)
- Shenbin Xu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jianan Lu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - John H Zhang
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA, United States.,Department of Anesthesiology, School of Medicine, Loma Linda University, Loma Linda, CA, United States.,Department of Neurosurgery, School of Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Jianmin Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Brain Research Institute, Zhejiang University, Hangzhou, China.,Collaborative Innovation Center for Brain Science, Zhejiang University, Hangzhou, China
| |
Collapse
|
42
|
Martha SR, Cheng Q, Fraser JF, Gong L, Collier LA, Davis SM, Lukins D, Alhajeri A, Grupke S, Pennypacker KR. Expression of Cytokines and Chemokines as Predictors of Stroke Outcomes in Acute Ischemic Stroke. Front Neurol 2020; 10:1391. [PMID: 32010048 PMCID: PMC6974670 DOI: 10.3389/fneur.2019.01391] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 12/18/2019] [Indexed: 12/30/2022] Open
Abstract
Introduction: Ischemic stroke remains one of the most debilitating diseases and is the fifth leading cause of death in the US. The ability to predict stroke outcomes within the acute period of stroke would be essential for care planning and rehabilitation. The Blood and Clot Thrombectomy Registry and Collaboration (BACTRAC; clinicaltrials.gov NCT03153683) study collects arterial blood immediately distal and proximal to the intracranial thrombus at the time of mechanical thrombectomy. These blood samples are an innovative resource in evaluating acute gene expression changes at the time of ischemic stroke. The purpose of this study was to identify inflammatory genes and important immune factors during mechanical thrombectomy for emergent large vessel occlusion (ELVO) and which patient demographics were predictors for stroke outcomes (infarct and/or edema volume) in acute ischemic stroke patients. Methods: The BACTRAC study is a non-probability sampling of male and female subjects (≥18 year old) treated with mechanical thrombectomy for ELVO. We evaluated 28 subjects (66 ± 15.48 years) relative concentrations of mRNA for gene expression in 84 inflammatory molecules in arterial blood distal and proximal to the intracranial thrombus who underwent thrombectomy. We used the machine learning method, Random Forest to predict which inflammatory genes and patient demographics were important features for infarct and edema volumes. To validate the overlapping genes with outcomes, we perform ordinary least squares regression analysis. Results: Machine learning analyses demonstrated that the genes and subject factors CCR4, IFNA2, IL-9, CXCL3, Age, T2DM, IL-7, CCL4, BMI, IL-5, CCR3, TNFα, and IL-27 predicted infarct volume. The genes and subject factor IFNA2, IL-5, CCL11, IL-17C, CCR4, IL-9, IL-7, CCR3, IL-27, T2DM, and CSF2 predicted edema volume. The overlap of genes CCR4, IFNA2, IL-9, IL-7, IL-5, CCR3, and IL-27 with T2DM predicted both infarct and edema volumes. These genes relate to a microenvironment for chemoattraction and proliferation of autoimmune cells, particularly Th2 cells and neutrophils. Conclusions: Machine learning algorithms can be employed to develop prognostic predictive biomarkers for stroke outcomes in ischemic stroke patients, particularly in regard to identifying acute gene expression changes that occur during stroke.
Collapse
Affiliation(s)
- Sarah R Martha
- School of Nursing, University of Washington, Seattle, WA, United States
| | - Qiang Cheng
- Institute for Biomedical Informatics, University of Kentucky, Lexington, KY, United States
| | - Justin F Fraser
- Department of Neurology, University of Kentucky, Lexington, KY, United States.,College of Medicine, University of Kentucky, Lexington, KY, United States.,Departments of Neurosurgery, University of Kentucky, Lexington, KY, United States.,Neuroscience, University of Kentucky, Lexington, KY, United States.,Radiology, University of Kentucky, Lexington, KY, United States
| | - Liyu Gong
- Institute for Biomedical Informatics, University of Kentucky, Lexington, KY, United States
| | - Lisa A Collier
- Department of Neurology, University of Kentucky, Lexington, KY, United States
| | - Stephanie M Davis
- Department of Neurology, University of Kentucky, Lexington, KY, United States
| | - Doug Lukins
- College of Medicine, University of Kentucky, Lexington, KY, United States.,Departments of Neurosurgery, University of Kentucky, Lexington, KY, United States.,Neuroscience, University of Kentucky, Lexington, KY, United States.,Radiology, University of Kentucky, Lexington, KY, United States
| | - Abdulnasser Alhajeri
- Department of Neurology, University of Kentucky, Lexington, KY, United States.,Radiology, University of Kentucky, Lexington, KY, United States
| | - Stephen Grupke
- Departments of Neurosurgery, University of Kentucky, Lexington, KY, United States.,Radiology, University of Kentucky, Lexington, KY, United States
| | - Keith R Pennypacker
- Department of Neurology, University of Kentucky, Lexington, KY, United States.,Neuroscience, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
43
|
Evaluation of sex differences in acid/base and electrolyte concentrations in acute large vessel stroke. Exp Neurol 2020; 323:113078. [DOI: 10.1016/j.expneurol.2019.113078] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 08/26/2019] [Accepted: 10/01/2019] [Indexed: 12/26/2022]
|
44
|
Li M, Cui MM, Kenechukwu NA, Gu YW, Chen YL, Zhong SJ, Gao YT, Cao XY, Wang L, Liu FM, Wen XR. Rosmarinic acid ameliorates hypoxia/ischemia induced cognitive deficits and promotes remyelination. Neural Regen Res 2020; 15:894-902. [PMID: 31719255 PMCID: PMC6990785 DOI: 10.4103/1673-5374.268927] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Rosmarinic acid, a common ester extracted from Rosemary, Perilla frutescens, and Salvia miltiorrhiza Bunge, has been shown to have protective effects against various diseases. This is an investigation into whether rosmarinic acid can also affect the changes of white matter fibers and cognitive deficits caused by hypoxic injury. The right common carotid artery of 3-day-old rats was ligated for 2 hours. The rats were then prewarmed in a plastic container with holes in the lid, which was placed in 37°C water bath for 30 minutes. Afterwards, the rats were exposed to an atmosphere with 8% O2 and 92% N2 for 30 minutes to establish the perinatal hypoxia/ischemia injury models. The rat models were intraperitoneally injected with rosmarinic acid 20 mg/kg for 5 consecutive days. At 22 days after birth, rosmarinic acid was found to improve motor, anxiety, learning and spatial memory impairments induced by hypoxia/ischemia injury. Furthermore, rosmarinic acid promoted the proliferation of oligodendrocyte progenitor cells in the subventricular zone. After hypoxia/ischemia injury, rosmarinic acid reversed to some extent the downregulation of myelin basic protein and the loss of myelin sheath in the corpus callosum of white matter structure. Rosmarinic acid partially slowed down the expression of oligodendrocyte marker Olig2 and myelin basic protein and the increase of oligodendrocyte apoptosis marker inhibitors of DNA binding 2. These data indicate that rosmarinic acid ameliorated the cognitive dysfunction after perinatal hypoxia/ischemia injury by improving remyelination in corpus callosum. This study was approved by the Animal Experimental Ethics Committee of Xuzhou Medical University, China (approval No. 20161636721) on September 16, 2017.
Collapse
Affiliation(s)
- Man Li
- Department of Genetics, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Miao-Miao Cui
- Department of Genetics, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | | | - Yi-Wei Gu
- Department of Urology, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Yu-Lin Chen
- Department of Genetics, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Si-Jing Zhong
- Xuzhou Medical University Clinical Medical College, Xuzhou, Jiangsu Province, China
| | - Yu-Ting Gao
- Department of Clinical Laboratory, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Xue-Yan Cao
- Department of Urology, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Li Wang
- Department of Urology, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Fu-Min Liu
- Department of Genetics, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Xiang-Ru Wen
- Research Center for Neurobiology and Department of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| |
Collapse
|
45
|
Orem BC, Partain SB, Stirling DP. Inhibiting store-operated calcium entry attenuates white matter secondary degeneration following SCI. Neurobiol Dis 2019; 136:104718. [PMID: 31846736 DOI: 10.1016/j.nbd.2019.104718] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 11/22/2019] [Accepted: 12/13/2019] [Indexed: 01/11/2023] Open
Abstract
Axonal degeneration plays a key role in the pathogenesis of numerous neurological disorders including spinal cord injury. After the irreversible destruction of the white matter elements during the primary (mechanical) injury, spared axons and their supporting glial cells begin to breakdown causing an expansion of the lesion site. Here we mechanistically link external sources of calcium entry through axoplasmic reticulum calcium store depletion that contributes to secondary axonal degeneration through a process called store-operated calcium entry. There is increasing evidence suggesting that store-operated calcium entry impairment is responsible for numerous disorders. Nevertheless, its role following spinal cord injury remains poorly understood. We hypothesize that store-operated calcium entry mediates secondary white matter degeneration after spinal cord injury. We used our previously published model of laser-induced spinal cord injury to focally transect mid cervical dorsal column axons from live 6-8-week-old heterozygous CNPaseGFP/+: Thy1YFP+ double transgenic murine spinal cord preparations (five treated, eight controls) and documented the dynamic changes in axons over time using two-photon excitation microscopy. We report that 1 hour delayed treatment with YM-58483, a potent inhibitor of store-operated calcium entry, significantly decreased intra-axonal calcium accumulation, axonal dieback both proximal and distal to the lesion site, reduced secondary axonal "bystander" damage acutely after injury, and promoted greater oligodendrocyte survival compared to controls. We also targeted store-operated calcium entry following a clinically relevant contusion spinal cord injury model in vivo. Adult, 6-8-week-old Advillin-Cre: Ai9 mice were subjected to a mild 30 kdyn contusion and imaged to observe secondary axonal degeneration in live animals. We found that delayed treatment with YM-58483 increased axonal survival and reduced axonal spheroid formation compared to controls (n = 5 mice per group). These findings suggest that blocking store-operated calcium entry acutely is neuroprotective and introduces a novel target to prevent pathological calcium entry following spinal cord injury using a clinically relevant model.
Collapse
Affiliation(s)
- Ben C Orem
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, KY 40202, USA; Department of Anatomical Sciences and Neurobiology, University of Louisville, Louisville, KY 40202, USA
| | - Steven B Partain
- Department of Bioengineering, University of Louisville, Louisville, KY 40202, USA
| | - David P Stirling
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, KY 40202, USA; Department of Anatomical Sciences and Neurobiology, University of Louisville, Louisville, KY 40202, USA; Department of Neurological Surgery, University of Louisville, Louisville, KY 40202, USA; Department of Microbiology and Immunology, University of Louisville, Louisville, KY 40202, USA.
| |
Collapse
|
46
|
Tamoxifen promotes white matter recovery and cognitive functions in male mice after chronic hypoperfusion. Neurochem Int 2019; 131:104566. [PMID: 31593788 DOI: 10.1016/j.neuint.2019.104566] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 09/30/2019] [Accepted: 10/02/2019] [Indexed: 12/18/2022]
Abstract
Cerebral white matter lesions (WMLs) induced by chronic cerebral hypoperfusion are one of the major components of stroke pathology and closely associated with cognitive impairment. However, the repair and related pathophysiology of white matter after brain injury remains relatively elusive and underexplored. Successful neuroregeneration is a method for the potential treatment of central nervous system (CNS) disorders. A non-steroidal estrogen receptor modulator, Tamoxifen, is an effective inhibitor of cell-swelling-activated anion channels and can mimic neuroprotective effects of estrogen in experimental ischemic stroke. However, its remains unclear whether Tamoxifen has beneficial effects in the pathological process after WMLs. In the present study, we investigated the efficacy of Tamoxifen on multiple elements of oligovascular niche of the male C57BL/6 mice brain after bilateral carotid artery stenosis (BCAS) - induced WMLs. Tamoxifen was injected intraperitoneally once daily from 1 day after BCAS until 1 day before sacrificed. Following chronic hypoperfusion, BCAS mice presented white matter demyelination, loss of axon-glia integrity, activated inflammatory response, and cognitive impairments. Tamoxifen treatment significantly facilitated functional restoration of working memory impairment in mice after white matter injury, thus indicating a translational potential for this estrogen receptor modulator given its clinical safety and applicability for WMLs, which lack of currently available treatments. Furthermore, Tamoxifen treatment reduced microglia activation and inflammatory response, favored microglial polarization toward to the M2 phenotype, enhanced oligodendrocyte precursor cells proliferation and differentiation, and promoted remyelination after chronic hypoperfusion. Together, our data indicate that Tamoxifen could alleviate white matter injury and play multiple targets protective effects following chronic hypoperfusion, which is a promising candidate for the therapeutic target for ischemic WMLs and other demyelination diseases associated cognitive impairment.
Collapse
|
47
|
Martha SR, Fraser JF, Pennypacker KR. Acid-Base and Electrolyte Changes Drive Early Pathology in Ischemic Stroke. Neuromolecular Med 2019; 21:540-545. [PMID: 31280473 DOI: 10.1007/s12017-019-08555-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 07/01/2019] [Indexed: 12/19/2022]
Abstract
Emergent large vessel occlusion accounts for 20-40% of ischemic strokes and is the most debilitating form of stroke. Some of the earliest changes in response to ischemic stroke occur in blood gases and electrolytes. These biochemical changes occur within minutes after occlusion in experimental models of stroke and can be utilized to predict stroke outcomes. The majority of ELVO stroke patients are middle-aged to elderly and are of both sexes, revealing that there is an age and sex mismatch between ischemic stroke patients and animal models, since most experimental studies use young male rats. Rethinking of the animal models should be considered, especially in encouraging the use of aged male and female rats with comorbidities to more closely mirror human populations. Mechanical thrombectomy provides a unique opportunity for researchers to further this work by expanding the collection and analysis of blood samples that are adjacent to the thrombus. To understand the complexity of stroke, researchers can analyze these tissues for different molecular targets that occur in response to ischemic stroke. This information may aid in the reduction of symptom burden for individuals diagnosed with ischemic stroke. Investigators should also focus on data from ischemic stroke patients and attempt to discover target molecules and then in animal models to establish mechanism, which will aid in the development of new stroke therapies. This review discusses the translation of these studies to the human patient to develop the capability to predict stroke outcomes. Future studies are needed to identify molecular targets to predict the risk of worsened long-term outcomes and/or increased risk for mortality.
Collapse
Affiliation(s)
- Sarah R Martha
- College of Nursing, University of Kentucky, Lexington, KY, USA
| | - Justin F Fraser
- Department of Neurology, University of Kentucky, Center for Advanced Translational Stroke Science, Building BBSRB, Office B377, Lexington, KY, 40536, USA.,Department of Neurosurgery, University of Kentucky, Lexington, KY, USA.,Department of Neuroscience, University of Kentucky, Lexington, KY, USA.,Department of Radiology, University of Kentucky, Lexington, KY, USA
| | - Keith R Pennypacker
- Department of Neurology, University of Kentucky, Center for Advanced Translational Stroke Science, Building BBSRB, Office B377, Lexington, KY, 40536, USA. .,Department of Neuroscience, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
48
|
Vaes JEG, Vink MA, de Theije CGM, Hoebeek FE, Benders MJNL, Nijboer CHA. The Potential of Stem Cell Therapy to Repair White Matter Injury in Preterm Infants: Lessons Learned From Experimental Models. Front Physiol 2019; 10:540. [PMID: 31143126 PMCID: PMC6521595 DOI: 10.3389/fphys.2019.00540] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 04/17/2019] [Indexed: 12/12/2022] Open
Abstract
Diffuse white matter injury (dWMI) is a major cause of morbidity in the extremely preterm born infant leading to life-long neurological impairments, including deficits in cognitive, motor, sensory, psychological, and behavioral functioning. At present, no treatment options are clinically available to combat dWMI and therefore exploration of novel strategies is urgently needed. In recent years, the pathophysiology underlying dWMI has slowly started to be unraveled, pointing towards the disturbed maturation of oligodendrocytes (OLs) as a key mechanism. Immature OL precursor cells in the developing brain are believed to be highly sensitive to perinatal inflammation and cerebral oxygen fluctuations, leading to impaired OL differentiation and eventually myelination failure. OL lineage development under normal and pathological circumstances and the process of (re)myelination have been studied extensively over the years, often in the context of other adult and pediatric white matter pathologies such as stroke and multiple sclerosis (MS). Various studies have proposed stem cell-based therapeutic strategies to boost white matter regeneration as a potential strategy against a wide range of neurological diseases. In this review we will discuss experimental studies focusing on mesenchymal stem cell (MSC) therapy to reduce white matter injury (WMI) in multiple adult and neonatal neurological diseases. What lessons have been learned from these previous studies and how can we translate this knowledge to application of MSCs for the injured white matter in the preterm infant? A perspective on the current state of stem cell therapy will be given and we will discuss different important considerations of MSCs including cellular sources, timing of treatment and administration routes. Furthermore, we reflect on optimization strategies that could potentially reinforce stem cell therapy, including preconditioning and genetic engineering of stem cells or using cell-free stem cell products, to optimize cell-based strategy for vulnerable preterm infants in the near future.
Collapse
Affiliation(s)
- Josine E G Vaes
- NIDOD Laboratory, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Department of Neonatology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Marit A Vink
- NIDOD Laboratory, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Caroline G M de Theije
- NIDOD Laboratory, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Freek E Hoebeek
- NIDOD Laboratory, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Manon J N L Benders
- Department of Neonatology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Cora H A Nijboer
- NIDOD Laboratory, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
49
|
Martha SR, Collier LA, Davis SM, Goodwin SJ, Powell D, Lukins D, Fraser JF, Pennypacker KR. Early acid/base and electrolyte changes in permanent middle cerebral artery occlusion: Aged male and female rats. J Neurosci Res 2019; 98:179-190. [PMID: 30942522 DOI: 10.1002/jnr.24422] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 02/28/2019] [Accepted: 03/11/2019] [Indexed: 01/04/2023]
Abstract
BACKGROUND Early changes in acid/base and electrolyte concentrations could provide insights into the development of neuropathology at the onset of stroke. We evaluated associations between acid/base and electrolyte concentrations, and outcomes in permanent middle cerebral artery occlusion (pMCAO) model. METHODS 18-month-old male and female Sprague-Dawley rats underwent pMCAO. Pre-, post- (7 min after occlusion), and at 72 hr of pMCAO venous blood samples provided pH, carbon dioxide, oxygen, glucose, hematocrit, hemoglobin, and electrolyte values of ionized calcium, potassium, and sodium. Multiple linear regression determined predictors of infarct and edema volumes from these values, Kaplan-Meier curve analyzed morality between males and females at 72 hr, and a Cox regression model was used to determine predictors for mortality. RESULTS Analysis indicated significant differences in acid/base balance and electrolyte levels in aged rats not dependent on sex between the three time points in the pMCAO model. Changes in pH (from pre- to post and post- to 72 hr) and changes in sodium and ionized calcium (from post- to 72 hr) were predictors of infarct volume and edema volume, respectively. Cox Regression revealed there is a 3.25 times increased risk for mortality based on changes in bicarbonate (pre- to post-MCAO). CONCLUSIONS These early venous blood changes in acid/base balance and electrolytes can be used to predict stroke outcomes in our rat model of stroke. This study provides potential biomarkers to be examined in the human condition that could provide profound prognostic tools for stroke patients.
Collapse
Affiliation(s)
- Sarah R Martha
- College of Nursing, University of Kentucky, Lexington, Kentucky
| | - Lisa A Collier
- Department of Neurology, University of Kentucky, Lexington, Kentucky
| | - Stephanie M Davis
- Department of Neurology, University of Kentucky, Lexington, Kentucky
| | - Sarah J Goodwin
- Department of Neurology, University of Kentucky, Lexington, Kentucky
| | - David Powell
- Magnetic Resonance Imaging and Spectroscopy Center, University of Kentucky, Lexington, Kentucky.,Department of Biomedical Imaging, University of Kentucky, Lexington, Kentucky
| | - Doug Lukins
- Department of Neurosurgery, University of Kentucky, Lexington, Kentucky.,Department of Neuroscience, University of Kentucky, Lexington, Kentucky.,Department of Radiology, University of Kentucky, Lexington, Kentucky
| | - Justin F Fraser
- Department of Neurology, University of Kentucky, Lexington, Kentucky.,Department of Neurosurgery, University of Kentucky, Lexington, Kentucky.,Department of Neuroscience, University of Kentucky, Lexington, Kentucky.,Department of Radiology, University of Kentucky, Lexington, Kentucky
| | - Keith R Pennypacker
- Department of Neurology, University of Kentucky, Lexington, Kentucky.,Department of Neuroscience, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
50
|
He S, Liu R, Li B, Huang L, Fan W, Tembachako CR, Zheng X, Xiong X, Miyata M, Xu B, Li Y, Fang W. Propagermanium, a CCR2 inhibitor, attenuates cerebral ischemia/reperfusion injury through inhibiting inflammatory response induced by microglia. Neurochem Int 2019; 125:99-110. [PMID: 30794846 DOI: 10.1016/j.neuint.2019.02.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 02/03/2019] [Accepted: 02/16/2019] [Indexed: 01/22/2023]
Abstract
CCR2 could recruit immune cells migrating into brain after ischemic stroke. It is unclear whether and why Propagermanium (PG, a CCR2 inhibitor) is able to protect against ischemic injury. Middle cerebral artery occlusion (MCAO) and reperfusion injury in C57BL/6 J male mice were performed in vivo to mimic ischemic stroke. Cultured BV2 microglia exposed to oxygen and glucose deprivation (OGD)/reoxygenation injury, LPS or IL-4 incubation were served in vitro. TTC staining, neurological score, brain water content, and MRI scan were performed to evaluate stroke outcome. Real time PCR, ELISA, and immunofluorescence were used to estimate inflammatory cytokines expression and releasing. Western blot was utilized to detect pSTAT1/STAT1 expression. Compared with MCAO mice, PG treatment significantly reduced infarction size and brain edema, improved neurological behavior at 72 h after MCAO. For inflammatory response, PG treatment inhibited inflammatory cytokines releasing, such as TNF-α, IFN-γ, IL-1β, IL-6, IL-12, IL-17, and IL-23. Further studies indicated that PG treatment downregulated mRNA expression of pro-inflammatory iNOS and CD86, and inhibited CD16 expressed in microglia. In vitro, PG incubation inhibited BV2 polarized to pro-inflammatory phenotype through STAT1 downregulation, while had no obvious effect on anti-inflammatory phenotype. Our observations suggest that CCR2 inhibitor PG downregulated pro-inflammatory microglia polarization for decreasing pro-inflammatory microglia phenotype marker, and thereafter inhibited inflammatory responses after MCAO in a STAT1-dependent manner.
Collapse
Affiliation(s)
- Shucheng He
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 210009, China
| | - Rui Liu
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 210009, China
| | - Binbin Li
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 210009, China
| | - Liangliang Huang
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 210009, China
| | - Wenxiang Fan
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 210009, China
| | - Charmaine Ruvimbo Tembachako
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 210009, China
| | - Xiaoya Zheng
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xiaoxing Xiong
- Renmin Hospital of Wuhan University, Wuhan, Hubei, 430006, China
| | - Masaaki Miyata
- Department of Cardiovascular Medicine, Kagoshima City Hospital, Japan
| | - Baohui Xu
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Yunman Li
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 210009, China.
| | - Weirong Fang
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 210009, China.
| |
Collapse
|