1
|
Hannawi Y. Cerebral Small Vessel Disease: a Review of the Pathophysiological Mechanisms. Transl Stroke Res 2024; 15:1050-1069. [PMID: 37864643 DOI: 10.1007/s12975-023-01195-9] [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: 06/02/2023] [Revised: 06/02/2023] [Accepted: 09/18/2023] [Indexed: 10/23/2023]
Abstract
Cerebral small vessel disease (cSVD) refers to the age-dependent pathological processes involving the brain small vessels and leading to vascular cognitive impairment, intracerebral hemorrhage, and acute lacunar ischemic stroke. Despite the significant public health burden of cSVD, disease-specific therapeutics remain unavailable due to the incomplete understanding of the underlying pathophysiological mechanisms. Recent advances in neuroimaging acquisition and processing capabilities as well as findings from cSVD animal models have revealed critical roles of several age-dependent processes in cSVD pathogenesis including arterial stiffness, vascular oxidative stress, low-grade systemic inflammation, gut dysbiosis, and increased salt intake. These factors interact to cause a state of endothelial cell dysfunction impairing cerebral blood flow regulation and breaking the blood brain barrier. Neuroinflammation follows resulting in neuronal injury and cSVD clinical manifestations. Impairment of the cerebral waste clearance through the glymphatic system is another potential process that has been recently highlighted contributing to the cognitive decline. This review details these mechanisms and attempts to explain their complex interactions. In addition, the relevant knowledge gaps in cSVD mechanistic understanding are identified and a systematic approach to future translational and early phase clinical research is proposed in order to reveal new cSVD mechanisms and develop disease-specific therapeutics.
Collapse
Affiliation(s)
- Yousef Hannawi
- Division of Cerebrovascular Diseases and Neurocritical Care, Department of Neurology, The Ohio State University, 333 West 10th Ave, Graves Hall 3172C, Columbus, OH, 43210, USA.
| |
Collapse
|
2
|
Lu WM, Ji HN, Yang RH, Cheng KL, Yang XL, Zeng HL, Tao K, Yin DM, Wu DH. A rat model of cerebral small vascular disease induced by ultrasound and protoporphyrin. Biochem Biophys Res Commun 2024; 735:150451. [PMID: 39094233 DOI: 10.1016/j.bbrc.2024.150451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/20/2024] [Accepted: 07/24/2024] [Indexed: 08/04/2024]
Abstract
Cerebral small vascular disease (CSVD) has a high incidence worldwide, but its pathological mechanisms remain poorly understood due to the lack of proper animal models. The current animal models of CSVD have several limitations such as high mortality rates and large-sized lesions, and thus it is urgent to develop new animal models of CSVD. Ultrasound can activate protoporphyrin to produce reactive oxygen species in a liquid environment. Here we delivered protoporphyrin into cerebral small vessels of rat brain through polystyrene microspheres with a diameter of 15 μm, and then performed transcranial ultrasound stimulation (TUS) on the model rats. We found that TUS did not affect the large vessels or cause large infarctions in the brain of model rats. The mortality rates were also comparable between the sham and model rats. Strikingly, TUS induced several CSVD-like phenotypes such as cerebral microinfarction, white matter injuries and impaired integrity of endothelial cells in the model rats. Additionally, these effects could be alleviated by antioxidant treatment with N-acetylcysteine (NAC). As control experiments, TUS did not lead to cerebral microinfarction in the rat brain when injected with the polystyrene microspheres not conjugated with protoporphyrin. In sum, we generated a rat model of CSVD that may be useful for the mechanistic study and drug development for CSVD.
Collapse
Affiliation(s)
- Wen-Mei Lu
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China; Joint Center for Translational Medicine, Shanghai Fifth People's Hospital, Fudan University and School of Life Science, East China Normal University, Shanghai, China
| | - Hao-Nan Ji
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China; Joint Center for Translational Medicine, Shanghai Fifth People's Hospital, Fudan University and School of Life Science, East China Normal University, Shanghai, China
| | - Rui-Hao Yang
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Kai-Li Cheng
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Xiao-Li Yang
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Hu-Lie Zeng
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China
| | - Ke Tao
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, China.
| | - Dong-Min Yin
- Joint Center for Translational Medicine, Shanghai Fifth People's Hospital, Fudan University and School of Life Science, East China Normal University, Shanghai, China.
| | - Dan-Hong Wu
- Department of Neurology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China; Joint Center for Translational Medicine, Shanghai Fifth People's Hospital, Fudan University and School of Life Science, East China Normal University, Shanghai, China.
| |
Collapse
|
3
|
Jiang H, Zhang C, Lin M, Yin Y, Deng S, Liu W, Zhuo B, Tian G, Du Y, Meng Z. Deciphering the mechanistic impact of acupuncture on the neurovascular unit in acute ischemic stroke: Insights from basic research in a narrative review. Ageing Res Rev 2024; 101:102536. [PMID: 39384155 DOI: 10.1016/j.arr.2024.102536] [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: 08/18/2024] [Revised: 09/27/2024] [Accepted: 10/01/2024] [Indexed: 10/11/2024]
Abstract
Ischemic stroke(IS), a severe acute cerebrovascular disease, not only imposes a heavy economic burden on society but also presents numerous challenges in treatment. During the acute phase, while thrombolysis and thrombectomy serve as primary treatments, these approaches are restricted by a narrow therapeutic window. During rehabilitation, commonly used neuroprotective agents struggle with their low drug delivery efficiency and inadequate preclinical testing, and the long-term pharmacological and toxicity effects of nanomedicines remain undefined. Meanwhile, acupuncture as a therapeutic approach is widely acknowledged for its effectiveness in treating IS and has been recommended by the World Health Organization (WHO) as an alternative and complementary therapy, even though its exact mechanisms remain unclear. This review aims to summarize the known mechanisms of acupuncture and electroacupuncture (EA) in the treatment of IS. Research shows that acupuncture treatment mainly protects the neurovascular unit through five mechanisms: 1) reducing neuronal apoptosis and promoting neuronal repair and proliferation; 2) maintaining the integrity of the blood-brain barrier (BBB); 3) inhibiting the overactivation and polarization imbalance of microglia; 4) regulating the movement of vascular smooth muscle (VSM) cells; 5) promoting the proliferation of oligodendrocyte precursors. Through an in-depth analysis, this review reveals the multi-level, multi-dimensional impact of acupuncture treatment on the neurovascular unit (NVU) following IS, providing stronger evidence and a theoretical basis for its clinical application.
Collapse
Affiliation(s)
- Hailun Jiang
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Chao Zhang
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Mengxuan Lin
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Yu Yin
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shizhe Deng
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Wei Liu
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Bifang Zhuo
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Guang Tian
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Yuzheng Du
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| | - Zhihong Meng
- Department of Acupuncture, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China; Department of Acupuncture, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| |
Collapse
|
4
|
Zhou Y, Xue R, Li Y, Ran W, Chen Y, Luo Z, Zhang K, Zhang R, Wang J, Fang M, Chen C, Lou M. Impaired Meningeal Lymphatics and Glymphatic Pathway in Patients with White Matter Hyperintensity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402059. [PMID: 38704728 PMCID: PMC11234435 DOI: 10.1002/advs.202402059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 04/07/2024] [Indexed: 05/07/2024]
Abstract
White matter hyperintensity (WMH) represents a critical global medical concern linked to cognitive decline and dementia, yet its underlying mechanisms remain poorly understood. Here, humans are directly demonstrated that high WMH burden correlates with delayed drainage of meningeal lymphatic vessels (mLVs) and glymphatic pathway. Additionally, a longitudinal cohort study reveals that glymphatic dysfunction predicts WMH progression. Next, in a rat model of WMH, the presence of impaired lymphangiogenesis and glymphatic drainage is confirmed, followed by elevated microglial activation and white matter demyelination. Notably, enhancing meningeal lymphangiogenesis through adeno-associated virus delivery of vascular endothelial growth factor-C (VEGF-C) mitigates microglial gliosis and white matter demyelination. Conversely, blocking the growth of mLVs with a VEGF-C trap strategy exacerbates these changes. The findings highlight the role of mLVs and glymphatic pathway dysfunction in aggravating brain white matter injury, providing a potential novel strategy for WMH prevention and treatment.
Collapse
Affiliation(s)
- Ying Zhou
- Department of Neurology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Rui Xue
- Department of Neurology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Yifei Li
- Department of Neurology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Wang Ran
- Department of Neurology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Yuping Chen
- Department of Neurology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Zhongyu Luo
- Department of Neurology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Kemeng Zhang
- Department of Neurology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Ruoxia Zhang
- Department of Neurology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Junjun Wang
- Department of Neurology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Mengmeng Fang
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Cong Chen
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Min Lou
- Department of Neurology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| |
Collapse
|
5
|
Seitz-Holland J, Alemán-Gómez Y, Cho KIK, Pasternak O, Cleusix M, Jenni R, Baumann PS, Klauser P, Conus P, Hagmann P, Do KQ, Kubicki M, Dwir D. Matrix metalloproteinase 9 (MMP-9) activity, hippocampal extracellular free water, and cognitive deficits are associated with each other in early phase psychosis. Neuropsychopharmacology 2024; 49:1140-1150. [PMID: 38431757 PMCID: PMC11109110 DOI: 10.1038/s41386-024-01814-5] [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: 07/13/2023] [Revised: 12/18/2023] [Accepted: 01/29/2024] [Indexed: 03/05/2024]
Abstract
Increasing evidence points toward the role of the extracellular matrix, specifically matrix metalloproteinase 9 (MMP-9), in the pathophysiology of psychosis. MMP-9 is a critical regulator of the crosstalk between peripheral and central inflammation, extracellular matrix remodeling, hippocampal development, synaptic pruning, and neuroplasticity. Here, we aim to characterize the relationship between plasma MMP-9 activity, hippocampal microstructure, and cognition in healthy individuals and individuals with early phase psychosis. We collected clinical, blood, and structural and diffusion-weighted magnetic resonance imaging data from 39 individuals with early phase psychosis and 44 age and sex-matched healthy individuals. We measured MMP-9 plasma activity, hippocampal extracellular free water (FW) levels, and hippocampal volumes. We used regression analyses to compare MMP-9 activity, hippocampal FW, and volumes between groups. We then examined associations between MMP-9 activity, FW levels, hippocampal volumes, and cognitive performance assessed with the MATRICS battery. All analyses were controlled for age, sex, body mass index, cigarette smoking, and years of education. Individuals with early phase psychosis demonstrated higher MMP-9 activity (p < 0.0002), higher left (p < 0.05) and right (p < 0.05) hippocampal FW levels, and lower left (p < 0.05) and right (p < 0.05) hippocampal volume than healthy individuals. MMP-9 activity correlated positively with hippocampal FW levels (all participants and individuals with early phase psychosis) and negatively with hippocampal volumes (all participants and healthy individuals). Higher MMP-9 activity and higher hippocampal FW levels were associated with slower processing speed and worse working memory performance in all participants. Our findings show an association between MMP-9 activity and hippocampal microstructural alterations in psychosis and an association between MMP-9 activity and cognitive performance. Further, more extensive longitudinal studies should examine the therapeutic potential of MMP-9 modulators in psychosis.
Collapse
Affiliation(s)
- Johanna Seitz-Holland
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Yasser Alemán-Gómez
- Connectomics Lab, Department of Radiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Medical Image Analysis Laboratory, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Kang Ik K Cho
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ofer Pasternak
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Martine Cleusix
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Raoul Jenni
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Philipp S Baumann
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Paul Klauser
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Service of Child and Adolescent Psychiatry, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Philippe Conus
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Patric Hagmann
- Connectomics Lab, Department of Radiology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Kim Q Do
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Marek Kubicki
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Daniella Dwir
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
6
|
Xu H, Xiao H, Tang Q. Lipopolysaccharide-induced intestinal inflammation on AIM2-mediated pyroptosis in the brain of rats with cerebral small vessel disease. Exp Neurol 2024; 375:114746. [PMID: 38428714 DOI: 10.1016/j.expneurol.2024.114746] [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: 12/08/2023] [Revised: 02/06/2024] [Accepted: 02/24/2024] [Indexed: 03/03/2024]
Abstract
Cerebral small vessel disease (CSVD) is a cerebral vascular disease with insidious onset and poor clinical treatment effect, which is related to neuroinflammation. This study investigated whether lipopolysaccharide-induced intestinal inflammation enhanced the level of pyroptosis in the brain of rats with CSVD. The bilateral carotid artery occlusion (BCAO) model was selected as the object of study. Firstly, behavioral tests and Hematoxylin-eosin staining (HE staining) were performed to determine whether the model was successful, and then the AIM2 inflammasome and pyroptosis indexes (AIM2, ASC, Caspase-1, IL-1β, GSDMD, N-GSDMD) in the brain were detected by Western blotting and Immunohistochemistry (IHC). Finally, a single intraperitoneal injection of lipopolysaccharide (LPS) was used to induce intestinal inflammation in rats, the expression of GSDMD and N-GSDMD in the brain was analyzed by Western blotting and to see if pyroptosis caused by intestinal inflammation can be inhibited by Disulfiram, an inhibitor of pyroptosis. The results showed that the inflammatory response and pyroptosis mediated by the AIM2 inflammasome in BCAO rats were present in both brain and intestine. The expression of N-GSDMD, a key marker of pyroptosis, in the brain was significantly increased and inhibited by Disulfiram after LPS-induced enhancement of intestinal inflammation. This study shows that AIM2-mediated inflammasome activation and pyroptosis exist in both brain and intestine in the rat model of CSVD. The enhancement of intestinal inflammation will increase the level of pyroptosis in the brain. In the future, targeted regulation of the AIM2 inflammasome may become a new strategy for the clinical treatment of CSVD.
Collapse
Affiliation(s)
- Huiping Xu
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Han Xiao
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Qiqiang Tang
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.
| |
Collapse
|
7
|
Guo YS, Bi X. Enriched environment enhanced the astrocyte-derived BDNF and VEGF expression and alleviate white matter injuries of rats with ischemic stroke. Neurol Res 2024; 46:272-283. [PMID: 38145566 DOI: 10.1080/01616412.2023.2298136] [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: 05/04/2023] [Accepted: 12/17/2023] [Indexed: 12/27/2023]
Abstract
OBJECTIVES Numerous studies have shown that an enriched environment can promote ischemic stroke and improve cognitive function. In addition, white matter is closely related to cognitive function. The effects and mechanisms of the enriched environment on white matter recovery after stroke have not been elucidated. This study will analyse the effects of the enriched environment on white matter and cognitive function in the post-stroke brain from the perspective of astrocytes and their secretions. METHODS Stroke models were used for middle cerebral artery occlusion model. post-operative rats were divided into sham-operated, standard and enriched environment groups. The degree of cerebral infarction was assessed by TTC staining and the degree of white matter damage was assessed by Luxol-Fast Blue staining. The prognosis after stroke was assessed using the longa score and Morris water maze test. Western Blot and immunofluorescence were used to quantify and localize astrocytes and their associated secretory factors and myelin protein markers. RESULTS We found that ischemic stroke can cause severe demyelination. After EE treatment, there was a significant increase in cerebral remyelination and a significant improvement in neurological and cognitive functions. Astrocyte, BDNF, and VEGF expression were significantly higher than in rats in the standard circumstances of stroke model. CONCLUSION These data suggest that the enriched environment contributes to brain white matter recovery and improvement of cognitive function after stroke. The mechanism is related to astrocytes and their secretions. EE can activate astrocytes to secrete BDNF and VEGF, which may be crucial to promote white matter recovery.
Collapse
Affiliation(s)
- Yi-Sha Guo
- Department of Physical Therapy, Affiliated Yangzhi Rehabilitation Hospital of Tongji University, Shanghai, China
- Department of rehabilitation medicine, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Xia Bi
- Department of rehabilitation medicine, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| |
Collapse
|
8
|
Askew KE, Beverley J, Sigfridsson E, Szymkowiak S, Emelianova K, Dando O, Hardingham GE, Duncombe J, Hennessy E, Koudelka J, Samarasekera N, Salman RA, Smith C, Tavares AAS, Gomez‐Nicola D, Kalaria RN, McColl BW, Horsburgh K. Inhibiting CSF1R alleviates cerebrovascular white matter disease and cognitive impairment. Glia 2024; 72:375-395. [PMID: 37909242 PMCID: PMC10952452 DOI: 10.1002/glia.24481] [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: 06/02/2023] [Revised: 09/27/2023] [Accepted: 10/04/2023] [Indexed: 11/02/2023]
Abstract
White matter abnormalities, related to poor cerebral perfusion, are a core feature of small vessel cerebrovascular disease, and critical determinants of vascular cognitive impairment and dementia. Despite this importance there is a lack of treatment options. Proliferation of microglia producing an expanded, reactive population and associated neuroinflammatory alterations have been implicated in the onset and progression of cerebrovascular white matter disease, in patients and in animal models, suggesting that targeting microglial proliferation may exert protection. Colony-stimulating factor-1 receptor (CSF1R) is a key regulator of microglial proliferation. We found that the expression of CSF1R/Csf1r and other markers indicative of increased microglial abundance are significantly elevated in damaged white matter in human cerebrovascular disease and in a clinically relevant mouse model of chronic cerebral hypoperfusion and vascular cognitive impairment. Using the mouse model, we investigated long-term pharmacological CSF1R inhibition, via GW2580, and demonstrated that the expansion of microglial numbers in chronic hypoperfused white matter is prevented. Transcriptomic analysis of hypoperfused white matter tissue showed enrichment of microglial and inflammatory gene sets, including phagocytic genes that were the predominant expression modules modified by CSF1R inhibition. Further, CSF1R inhibition attenuated hypoperfusion-induced white matter pathology and rescued spatial learning impairments and to a lesser extent cognitive flexibility. Overall, this work suggests that inhibition of CSF1R and microglial proliferation mediates protection against chronic cerebrovascular white matter pathology and cognitive deficits. Our study nominates CSF1R as a target for the treatment of vascular cognitive disorders with broader implications for treatment of other chronic white matter diseases.
Collapse
Affiliation(s)
| | - Joshua Beverley
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
| | - Emma Sigfridsson
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
| | - Stefan Szymkowiak
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
- UK Dementia Research InstituteUniversity of EdinburghEdinburghUK
| | - Katherine Emelianova
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
- UK Dementia Research InstituteUniversity of EdinburghEdinburghUK
| | - Owen Dando
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
- UK Dementia Research InstituteUniversity of EdinburghEdinburghUK
| | - Giles E. Hardingham
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
- UK Dementia Research InstituteUniversity of EdinburghEdinburghUK
| | - Jessica Duncombe
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
| | - Edel Hennessy
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
| | - Juraj Koudelka
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
- UK Dementia Research InstituteUniversity of EdinburghEdinburghUK
| | - Neshika Samarasekera
- Centre for Clinical Brain Sciences and Sudden Death Brain BankUniversity of EdinburghEdinburghUK
| | - Rustam Al‐Shahi Salman
- Centre for Clinical Brain Sciences and Sudden Death Brain BankUniversity of EdinburghEdinburghUK
| | - Colin Smith
- Centre for Clinical Brain Sciences and Sudden Death Brain BankUniversity of EdinburghEdinburghUK
| | - Adriana A. S. Tavares
- British Heart Foundation Centre for Cardiovascular ScienceUniversity of EdinburghEdinburghUK
| | | | - Raj N. Kalaria
- Clinical and Translational Research InstituteNewcastle UniversityNewcastleUK
| | - Barry W. McColl
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
- UK Dementia Research InstituteUniversity of EdinburghEdinburghUK
| | - Karen Horsburgh
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
| |
Collapse
|
9
|
Zeng M, Peng M, Liang J, Sun H. The Role of Gut Microbiota in Blood-Brain Barrier Disruption after Stroke. Mol Neurobiol 2023:10.1007/s12035-023-03512-7. [PMID: 37498481 DOI: 10.1007/s12035-023-03512-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 07/13/2023] [Indexed: 07/28/2023]
Abstract
Growing evidence has proved that alterations in the gut microbiota have been linked to neurological disorders including stroke. Structural and functional disruption of the blood-brain barrier (BBB) is observed after stroke. In this context, there is pioneering evidence supporting that gut microbiota may be involved in the pathogenesis of stroke by regulating the BBB function. However, only a few experimental studies have been performed on stroke models to observe the BBB by altering the structure of gut microbiota, which warrant further exploration. Therefore, in order to provide a novel mechanism for stroke and highlight new insights into BBB modification as a stroke intervention, this review summarizes existing evidence of the relationship between gut microbiota and BBB integrity and discusses the mechanisms of gut microbiota on BBB dysfunction and its role in stroke.
Collapse
Affiliation(s)
- Meiqin Zeng
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, 510280, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China On Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory On Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Meichang Peng
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, 510280, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China On Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory On Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Jianhao Liang
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, 510280, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China On Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory On Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Haitao Sun
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, 510280, Guangzhou, China.
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China On Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory On Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China.
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Centre for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou, China.
| |
Collapse
|
10
|
Low vitamin D status is associated with inflammatory response in older patients with cerebral small vessel disease. J Neuroimmunol 2023; 377:578057. [PMID: 36921477 DOI: 10.1016/j.jneuroim.2023.578057] [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: 12/15/2022] [Revised: 02/18/2023] [Accepted: 02/25/2023] [Indexed: 03/11/2023]
Abstract
OBJECTIVES This study aimed to determine the association of the NF-κB inflammatory signaling pathway with vitamin D status in older cerebral small vessel disease (SVD) patients. METHODS We measured serum 25(OH)D, pro-and anti-inflammatory cytokines, and mRNA levels of the vitamin D-activating enzyme, CYP27B1, as well as NF-kB, COX-2, the chemokine-CCL2, IL-1β, IL-6, TNF-α, TGF-β, and IL-10, in cerebral SVD patients aged ≥60 years presenting with vascular dementia and age and gender-matched healthy controls. RESULTS Low vitamin D status (insufficiency: serum 25(OH)D 12-20 ng/ml; deficiency: ≤12 ng/ml) was more prevalent among patients compared to controls. The mRNA levels of NF-kB, COX-2, CCL2, IL-1β, and IL-6, and serum levels of pro-inflammatory cytokines (IL-1α, IL-1β, IL-6, and TNF-α) were significantly higher in cases compared to controls. There was a significant correlation between CYP27B1 and NF-kB, COX-2, CCL2, and IL-1β gene expression. Serum IL-1α, IL-1β, and IL-6 concentrations and the expression of CCL-2, NF-kB2, and NF-kB3 genes were higher in vitamin D-deficient subjects compared to vitamin D-sufficient subjects. There was a significant negative correlation between serum 25(OH)D and IL-1α, IL-6, and TNF-α, and a positive correlation between 25(OH)D and IL-10. CONCLUSION Low vitamin D is associated with an inflammatory response via NF-kB signaling, which could play a role in the etio-pathogenesis of SVD. Further large-scale studies are required to validate our findings.
Collapse
|
11
|
Dobrynina LA, Makarova AG, Shabalina AA, Burmak AG, Shlapakova PS, Shamtieva KV, Tsypushtanova MM, Trubitsyna VV, Gnedovskaya EV. [A role of altered inflammation-related gene expression in cerebral small vessel disease with cognitive impairment]. Zh Nevrol Psikhiatr Im S S Korsakova 2023; 123:58-68. [PMID: 37796069 DOI: 10.17116/jnevro202312309158] [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: 10/06/2023]
Abstract
OBJECTIVE To identify the role of changes in the expression of inflammation-related genes in cerebral microangiopathy/cerebral small vessel disease (cSVD). MATERIAL AND METHODS Forty-four cSVD patients (mean age 61.4±9.2) and 11 controls (mean age 57.3±9.7) were studied. Gene expression was assessed on an individual NanoString nCounter panel of 58 inflammation-related genes and 4 reference genes. A set of genes was generated based on converging results of complete genome-wide association studies (GWAS) in cSVD and Alzheimer's disease (AD) and circulating markers associated with vascular wall and Brain lesions in cSVD. RNA was isolated from blood leukocytes and analyzed with the nCounter Analysis System, followed by analysis in nSolver 4.0. Results were verified by real-time PCR. RESULTS CSVD patients had a significant decrease in BIN1 (log2FC=-1.272; p=0.039) and VEGFA (log2FC=-1.441; p=0.038) expression compared to controls, which showed predictive ability for cSVD. The cut-off for BIN1 expression was 5.76 a.u. (sensitivity 73%; specificity 75%) and the cut-off for VEGFA expression was 9.27 a.u. (sensitivity 64%; specificity 86%). Reduced expression of VEGFA (p=0.011), VEGFC (p=0.017), CD2AP (p=0.044) was associated with cognitive impairment (CI). There was a significant direct correlation between VEGFC expression and the scores on the Montreal Cognitive Assessment test and between BIN1 and VEGFC expression and delayed memory. CONCLUSION The possible prediction of cSVD by reduced expression levels of BIN1, VEGFA and the association of clinically significant CI with reduced VEGFA and VEGFC expression indicate their importance in the development and progression of the disease. The established importance of these genes in the pathogenesis of AD suggests that similar changes in their expression profile in cSVD may be one of the conditions for the comorbidity of the two pathologies.
Collapse
Affiliation(s)
| | | | | | - A G Burmak
- Research Center of Neurology, Moscow, Russia
| | | | | | | | | | | |
Collapse
|
12
|
Rosenberg GA. Willis Lecture: Biomarkers for Inflammatory White Matter Injury in Binswanger Disease Provide Pathways to Precision Medicine. Stroke 2022; 53:3514-3523. [PMID: 36148658 PMCID: PMC9613611 DOI: 10.1161/strokeaha.122.039211] [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: 11/16/2022]
Abstract
Binswanger disease is the small vessel form of vascular cognitive impairment and dementia. Deposition of Alzheimer disease proteins can begin in midlife and progress slowly, whereas aging of the vasculature also can begin in midlife, continuing to progress into old age, making mixed dementia the most common type of dementia. Biomarkers facilitate the early diagnosis of dementias. It is possible to diagnose mixed dementia before autopsy with biomarkers for vascular disease derived from diffusor tensor images on magnetic resonance imaging and Alzheimer disease proteins, Aβ (amyloid β), and phosphorylated tau, in cerebrospinal fluid or in brain with positron emission tomography. The presence of vascular disease accelerates cognitive decline. Both misfolded proteins and vascular disease promote inflammation, which can be detected in cerebrospinal fluid by the presence of MMPs (matrix metalloproteinases), angiogenic growth factors, and cytokines. MMPs disrupt the blood-brain barrier and break down myelin, producing Binswanger disease's 2 main pathological features. Advances in detecting biomarkers in plasma will provide early detection of dementia and aided by machine learning and artificial intelligence, will enhance diagnosis and form the basis for early treatments.
Collapse
Affiliation(s)
- Gary A Rosenberg
- Center for Memory and Aging, Departments of Neurology, Neurosciences, Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque
| |
Collapse
|
13
|
Ji X, Tian L, Niu S, Yao S, Qu C. Trimethylamine N-oxide promotes demyelination in spontaneous hypertension rats through enhancing pyroptosis of oligodendrocytes. Front Aging Neurosci 2022; 14:963876. [PMID: 36072486 PMCID: PMC9441869 DOI: 10.3389/fnagi.2022.963876] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 07/29/2022] [Indexed: 12/24/2022] Open
Abstract
Background Hypertension is a leading risk factor for cerebral small vessel disease (CSVD), a brain microvessels dysfunction accompanied by white matter lesions (WML). Trimethylamine N-oxide (TMAO), a metabolite of intestinal flora, is correlated with cardiovascular and aging diseases. Here, we explored the effect of TMAO on the demyelination of WML. Methods Spontaneous hypertension rats (SHRs) and primary oligodendrocytes were used to explore the effect of TMAO on demyelination in vivo and in vitro. T2-weighted magnetic resonance imaging (MRI) was applied to characterize the white matter hyperintensities (WMH) in rats. TMAO level was evaluated using LC-MS/MS assay. The histopathological changes of corpus callosum were measured by hematoxylin-eosin and luxol fast blue staining. And the related markers were detected by IHC, IF and western blot assay. Mito Tracker Red probe, DCFH-DA assay, flow cytometry based on JC-1 staining and Annexin V-FITC/PI double staining were conducted to evaluate the mitochondrial function, intracellular ROS levels and cell apoptosis. Results SHRs exhibited stronger WMH signals and a higher TMAO level than age-matched normotensive Wistar-kyoto rats (WKY). The corpus callosum region of SHR showed decreased volumes and enhanced demyelination when treated with TMAO. Furthermore, TMAO significantly elevated ROS production and induced NLRP3 inflammasome and impairment of mitochondrial function of oligodendrocytes. More importantly, TMAO enhanced the pyroptosis-related inflammatory death of oligodendrocytes. Conclusion TMAO could cross the blood-brain barrier (BBB) and promote oligodendrocytes pyroptosis via ROS/NLRP3 inflammasome signaling and mitochondrial dysfunction to promote demyelination, revealing a new diagnostic marker for WML under hypertension.
Collapse
Affiliation(s)
- Xiaotan Ji
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, China
- Department of Neurology, Jining No. 1 People’s Hospital, Jining, China
| | - Long Tian
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Shenna Niu
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Shumei Yao
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Chuanqiang Qu
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, China
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- *Correspondence: Chuanqiang Qu,
| |
Collapse
|
14
|
Gao Y, Li D, Lin J, Thomas AM, Miao J, Chen D, Li S, Chu C. Cerebral small vessel disease: Pathological mechanisms and potential therapeutic targets. Front Aging Neurosci 2022; 14:961661. [PMID: 36034144 PMCID: PMC9412755 DOI: 10.3389/fnagi.2022.961661] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 07/29/2022] [Indexed: 11/13/2022] Open
Abstract
Cerebral small vessel disease (CSVD) represents a diverse cluster of cerebrovascular diseases primarily affecting small arteries, capillaries, arterioles and venules. The diagnosis of CSVD relies on the identification of small subcortical infarcts, lacunes, white matter hyperintensities, perivascular spaces, and microbleeds using neuroimaging. CSVD is observed in 25% of strokes worldwide and is the most common pathology of cognitive decline and dementia in the elderly. Still, due to the poor understanding of pathophysiology in CSVD, there is not an effective preventative or therapeutic approach for CSVD. The most widely accepted approach to CSVD treatment is to mitigate vascular risk factors and adopt a healthier lifestyle. Thus, a deeper understanding of pathogenesis may foster more specific therapies. Here, we review the underlying mechanisms of pathological characteristics in CSVD development, with a focus on endothelial dysfunction, blood-brain barrier impairment and white matter change. We also describe inflammation in CSVD, whose role in contributing to CSVD pathology is gaining interest. Finally, we update the current treatments and preventative measures of CSVD, as well as discuss potential targets and novel strategies for CSVD treatment.
Collapse
Affiliation(s)
- Yue Gao
- Department of Neurointervention and Neurological Intensive Care, Dalian Municipal Central Hospital, Dalian, China
| | - Di Li
- Department of Neurointervention and Neurological Intensive Care, Dalian Municipal Central Hospital, Dalian, China
| | - Jianwen Lin
- Department of Neurology, Dalian Municipal Central Hospital, Dalian, China
| | - Aline M. Thomas
- F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institution, Baltimore, MD, United States
| | - Jianyu Miao
- Department of Neurology, Dalian Municipal Central Hospital, Dalian, China
| | - Dong Chen
- Department of Neurosurgery, Dalian Municipal Central Hospital, Dalian, China
| | - Shen Li
- Department of Neurology and Psychiatry, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Chengyan Chu
- Department of Neurology, Dalian Municipal Central Hospital, Dalian, China
- *Correspondence: Chengyan Chu,
| |
Collapse
|
15
|
Jenkins TA. Metabolic Syndrome and Vascular-Associated Cognitive Impairment: a Focus on Preclinical Investigations. Curr Diab Rep 2022; 22:333-340. [PMID: 35737273 PMCID: PMC9314301 DOI: 10.1007/s11892-022-01475-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/22/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE OF REVIEW Metabolic syndrome is associated with an increased risk of vascular cognitive impairment or, in the more extreme, vascular dementia. Animal models are used to investigate the relationship between pathology and behaviour. This review summarizes the latest understanding of the role of the hippocampus and prefrontal cortex in vascular cognitive impairment, the influence of inflammation in this association while also commenting on some of the latest interventions proposed. RECENT FINDINGS Models of vascular cognitive impairment and vascular dementia, whether they develop from an infarct or non-infarct base, demonstrate increased neuroinflammation, reduced neuronal function and deficits in prefrontal and hippocampal-associated cognitive domains. Promising new research shows agents and environmental interventions that inhibit central oxidative stress and inflammation can reverse both pathology and cognitive dysfunction. While preclinical studies suggest that reversal of deficits in vascular cognitive impairment models is possible, replication in patients still needs to be demonstrated.
Collapse
Affiliation(s)
- Trisha A Jenkins
- Human Biosciences, School of Health and Biomedical Sciences, STEM College, RMIT University, Plenty Road, Bundoora, VIC, 3083, Australia.
| |
Collapse
|
16
|
The Underlying Role of the Glymphatic System and Meningeal Lymphatic Vessels in Cerebral Small Vessel Disease. Biomolecules 2022; 12:biom12060748. [PMID: 35740873 PMCID: PMC9221030 DOI: 10.3390/biom12060748] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/21/2022] [Accepted: 05/24/2022] [Indexed: 02/01/2023] Open
Abstract
There is a growing prevalence of vascular cognitive impairment (VCI) worldwide, and most research has suggested that cerebral small vessel disease (CSVD) is the main contributor to VCI. Several potential physiopathologic mechanisms have been proven to be involved in the process of CSVD, such as blood-brain barrier damage, small vessels stiffening, venous collagenosis, cerebral blood flow reduction, white matter rarefaction, chronic ischaemia, neuroinflammation, myelin damage, and subsequent neurodegeneration. However, there still is a limited overall understanding of the sequence and the relative importance of these mechanisms. The glymphatic system (GS) and meningeal lymphatic vessels (mLVs) are the analogs of the lymphatic system in the central nervous system (CNS). As such, these systems play critical roles in regulating cerebrospinal fluid (CSF) and interstitial fluid (ISF) transport, waste clearance, and, potentially, neuroinflammation. Accumulating evidence has suggested that the glymphatic and meningeal lymphatic vessels played vital roles in animal models of CSVD and patients with CSVD. Given the complexity of CSVD, it was significant to understand the underlying interaction between glymphatic and meningeal lymphatic transport with CSVD. Here, we provide a novel framework based on new advances in main four aspects, including vascular risk factors, potential mechanisms, clinical subtypes, and cognition, which aims to explain how the glymphatic system and meningeal lymphatic vessels contribute to the progression of CSVD and proposes a comprehensive insight into the novel therapeutic strategy of CSVD.
Collapse
|
17
|
Garcia-Martin G, Alcover-Sanchez B, Wandosell F, Cubelos B. Pathways Involved in Remyelination after Cerebral Ischemia. Curr Neuropharmacol 2022; 20:751-765. [PMID: 34151767 PMCID: PMC9878953 DOI: 10.2174/1570159x19666210610093658] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 05/05/2021] [Accepted: 05/12/2021] [Indexed: 11/22/2022] Open
Abstract
Brain ischemia, also known as ischemic stroke, occurs when there is a lack of blood supply into the brain. When an ischemic insult appears, both neurons and glial cells can react in several ways that will determine the severity and prognosis. This high heterogeneity of responses has been a major obstacle in developing effective treatments or preventive methods for stroke. Although white matter pathophysiology has not been deeply assessed in stroke, its remodelling can greatly influence the clinical outcome and the disability degree. Oligodendrocytes, the unique cell type implied in CNS myelination, are sensible to ischemic damage. Loss of myelin sheaths can compromise axon survival, so new Oligodendrocyte Precursor Cells are required to restore brain function. Stroke can, therefore, enhance oligodendrogenesis to regenerate those new oligodendrocytes that will ensheath the damaged axons. Given that myelination is a highly complex process that requires coordination of multiple pathways such as Sonic Hedgehog, RTKs or Wnt/β-catenin, we will analyse new research highlighting their importance after brain ischemia. In addition, oligodendrocytes are not isolated cells inside the brain, but rather form part of a dynamic environment of interactions between neurons and glial cells. For this reason, we will put some context into how microglia and astrocytes react against stroke and influence oligodendrogenesis to highlight the relevance of remyelination in the ischemic brain. This will help to guide future studies to develop treatments focused on potentiating the ability of the brain to repair the damage.
Collapse
Affiliation(s)
- Gonzalo Garcia-Martin
- Departamento de Biología Molecular and Centro Biología Molecular “Severo Ochoa”, Universidad Autónoma de Madrid-Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain
| | - Berta Alcover-Sanchez
- Departamento de Biología Molecular and Centro Biología Molecular “Severo Ochoa”, Universidad Autónoma de Madrid-Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain
| | - Francisco Wandosell
- Departamento de Biología Molecular and Centro Biología Molecular “Severo Ochoa”, Universidad Autónoma de Madrid-Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain
| | - Beatriz Cubelos
- Departamento de Biología Molecular and Centro Biología Molecular “Severo Ochoa”, Universidad Autónoma de Madrid-Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain,Address correspondence to this author at the Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Nicolás Cabrera 1, Universidad Autónoma de Madrid, 28049 Madrid, Spain; Tel: 34-91-1964561; Fax: 34-91-1964420; E-mail:
| |
Collapse
|
18
|
Panda SP, Soni U. A review of dementia, focusing on the distinct roles of viral protein corona and MMP9 in dementia: Potential pharmacotherapeutic priorities. Ageing Res Rev 2022; 75:101560. [PMID: 35031512 DOI: 10.1016/j.arr.2022.101560] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 12/27/2021] [Accepted: 01/07/2022] [Indexed: 02/08/2023]
Abstract
Dementia, in particular, is a defining feature of Alzheimer's and Parkinson's diseases. Because of the combination of motor and cognitive impairments, Parkinson's disease dementia (PDD) has a greater impact on affected people than Alzheimer's disease dementia (ADD) and others. If one family member develops dementia, the other members will suffer greatly in terms of social and occupational functioning. Currently, no relevant treatment is available based on an examination of the absolute pathophysiology of dementia. As a result, our objective of current review encouraged to look for dementia pharmacotherapy based on their pathogenesis. We systematically searched electronic databases such as PubMed, Scopus, and ESCI for information on the pathophysiology of demetia, as well as their treatment with allopathic and herbal medications. By modulating intermediate proteins, oxidative stress, viral protein corona, and MMP9 are etiological factors that cause dementia. The pathophysiology of ADD was described by two hypotheses: the amyloid cascade hypothesis and the tau and tangle hypothesis. ADD is caused by an increase in amyloid-beta (Aβ) and neurofibrillary tangles in the cerebrum. The viral protein corona (VPC) is more contagious and helps to form amyloid-beta (Aβ) plaques and neurofibrillary tangles in the cerebrum. Thioredoxin interacting protein (TXNIP) inside the BBB encourages Aβ to become more engaged. PDD is caused by decreased or absent dopamine secretion from nerve cells in the substantia nigra, as well as PRKN gene deletion/duplication mutations, and shift in the PRKN-PACRG organisation, all of which are linked to ageing. This article discussed the pathophysiology of dementia, as well as a list of herbal medications that can easily cross the BBB and have a therapeutic effect on dementia.
Collapse
|
19
|
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: 8.5] [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
|
20
|
Gliovascular Mechanisms and White Matter Injury in Vascular Cognitive Impairment and Dementia. Stroke 2022. [DOI: 10.1016/b978-0-323-69424-7.00013-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
21
|
Li S, Li G, Luo X, Huang Y, Wen L, Li J. Endothelial Dysfunction and Hyperhomocysteinemia-Linked Cerebral Small Vessel Disease: Underlying Mechanisms and Treatment Timing. Front Neurol 2021; 12:736309. [PMID: 34899561 PMCID: PMC8651556 DOI: 10.3389/fneur.2021.736309] [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: 07/09/2021] [Accepted: 11/01/2021] [Indexed: 02/05/2023] Open
Abstract
Cerebral small vessel disease (cSVD)—a common cause of stroke and vascular dementia—is a group of clinical syndromes that affects the brain's small vessels, including arterioles, capillaries, and venules. Its pathogenesis is not fully understood, and effective treatments are limited. Increasing evidence indicates that an elevated total serum homocysteine level is directly and indirectly associated with cSVD, and endothelial dysfunction plays an active role in this association. Hyperhomocysteinemia affects endothelial function through oxidative stress, inflammatory pathways, and epigenetic alterations at an early stage, even before the onset of small vessel injuries and the disease. Therefore, hyperhomocysteinemia is potentially an important therapeutic target for cSVD. However, decreasing the homocysteine level is not sufficiently effective, possibly due to delayed treatment, which underlying reason remains unclear. In this review, we examined endothelial dysfunction to understand the close relationship between hyperhomocysteinemia and cSVD and identify the optimal timing for the therapy.
Collapse
Affiliation(s)
- Shuang Li
- Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Guangjian Li
- Department of Neurology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xia Luo
- Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yan Huang
- Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Lan Wen
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Jinglun Li
- Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
22
|
Evans LE, Taylor JL, Smith CJ, Pritchard HAT, Greenstein AS, Allan SM. Cardiovascular co-morbidities, inflammation and cerebral small vessel disease. Cardiovasc Res 2021; 117:2575-2588. [PMID: 34499123 DOI: 10.1093/cvr/cvab284] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Indexed: 12/15/2022] Open
Abstract
Cerebral small vessel disease (cSVD) is the most common cause of vascular cognitive impairment and affects all levels of the brain's vasculature. Features include diverse structural and functional changes affecting small arteries and capillaries that lead to a decline in cerebral perfusion. Due to an aging population, incidence of cerebral small vessel disease (cSVD) is continually rising. Despite its prevalence and its ability to cause multiple debilitating illnesses, such as stroke and dementia, there are currently no therapeutic strategies for the treatment of cSVD. In the healthy brain, interactions between neuronal, vascular and inflammatory cells are required for normal functioning. When these interactions are disturbed, chronic pathological inflammation can ensue. The interplay between cSVD and inflammation has attracted much recent interest and this review discusses chronic cardiovascular diseases, particularly hypertension, and explores how the associated inflammation may impact on the structure and function of the small arteries of the brain in cSVD. Molecular approaches in animal studies are linked to clinical outcomes in patients and novel hypotheses regarding inflammation and cSVD are proposed that will hopefully stimulate further discussion and study in this important area.
Collapse
Affiliation(s)
- Lowri E Evans
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom.,Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK
| | - Jade L Taylor
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom.,Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK
| | - Craig J Smith
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom.,Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK.,Manchester Centre for Clinical Neurosciences, Manchester Academic Health Science Centre, Salford Royal Hospital, Manchester Academic Health Sciences Centre (MAHSC)
| | - Harry A T Pritchard
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom.,Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK
| | - Adam S Greenstein
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom.,Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK
| | - Stuart M Allan
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK.,Division of Neuroscience and Experimental Psychology, The University of Manchester, Manchester, UK
| |
Collapse
|
23
|
Rastogi A, Weissert R, Bhaskar SMM. Emerging role of white matter lesions in cerebrovascular disease. Eur J Neurosci 2021; 54:5531-5559. [PMID: 34233379 DOI: 10.1111/ejn.15379] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/26/2021] [Accepted: 06/26/2021] [Indexed: 12/12/2022]
Abstract
White matter lesions have been implicated in the setting of stroke, dementia, intracerebral haemorrhage, several other cerebrovascular conditions, migraine, various neuroimmunological diseases like multiple sclerosis, disorders of metabolism, mitochondrial diseases and others. While much is understood vis a vis neuroimmunological conditions, our knowledge of the pathophysiology of these lesions, and their role in, and implications to, management of cerebrovascular diseases or stroke, especially in the elderly, are limited. Several clinical assessment tools are available for delineating white matter lesions in clinical practice. However, their incorporation into clinical decision-making and specifically prognosis and management of patients is suboptimal for use in standards of care. This article sought to provide an overview of the current knowledge and recent advances on pathophysiology, as well as clinical and radiological assessment, of white matter lesions with a focus on its development, progression and clinical implications in cerebrovascular diseases. Key indications for clinical practice and recommendations on future areas of research are also discussed. Finally, a conceptual proposal on putative mechanisms underlying pathogenesis of white matter lesions in cerebrovascular disease has been presented. Understanding of pathophysiology of white matter lesions and how they mediate outcomes is important to develop therapeutic strategies.
Collapse
Affiliation(s)
- Aarushi Rastogi
- South Western Sydney Clinical School, University of New South Wales (UNSW), Liverpool, New South Wales, Australia.,Neurovascular Imaging Laboratory, Clinical Sciences Stream, Ingham Institute for Applied Medical Research, Sydney, New South Wales, Australia
| | - Robert Weissert
- Department of Neurology, Regensburg University Hospital, University of Regensburg, Regensburg, Germany
| | - Sonu Menachem Maimonides Bhaskar
- South Western Sydney Clinical School, University of New South Wales (UNSW), Liverpool, New South Wales, Australia.,Neurovascular Imaging Laboratory, Clinical Sciences Stream, Ingham Institute for Applied Medical Research, Sydney, New South Wales, Australia.,NSW Brain Clot Bank, NSW Health Pathology, Sydney, New South Wales, Australia.,Department of Neurology and Neurophysiology, Liverpool Hospital and South Western Sydney Local Health District, Sydney, New South Wales, Australia
| |
Collapse
|
24
|
Qin M, Liu Y, Sun M, Li X, Xu J, Zhang L, Jiang H. Protective effects of melatonin on the white matter damage of neonatal rats by regulating NLRP3 inflammasome activity. Neuroreport 2021; 32:739-747. [PMID: 33994520 DOI: 10.1097/wnr.0000000000001642] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE The aim of the study was to investigate the protective effects and relevant mechanisms of melatonin on the white matter damage (WMD) caused by endotoxin and ischemic hypoxia in neonatal rats. METHODS Seventy-two female neonatal rats (postnatal day 3) were randomly divided into the sham, melatonin-treated, and control groups (n = 24 for each group). The periventricular white matter was collected to evaluate the WMD and apoptosis. In addition, the reactive oxygen species (ROS) level was measured. The expression levels of nucleotide-binding domain-like receptor protein 3 (NLRP3), interleukin (IL)-1β, IL-18, pink1, parkin, Toll-like receptor (TLR)-4, and nuclear factor (NF)-κB were detected. RESULTS Hematoxylin and eosin and terminal-deoxynucleoitidyl transferase mediated nick end labeling staining showed that the WMD, as well as cell degeneration, necrosis, and apoptosis in the control group, were more severe than those in the melatonin-treated group. Endotoxin and ischemic hypoxia upregulated the expression of NLRP3 and downstream inflammatory factors such as IL-1β and IL-18, which could be reversed by melatonin treatment. Melatonin increased mitochondrial autophagy marker (pink1 and parkin) expression in the white matter and reduced ROS production. Moreover, melatonin-reduced TLR4 and NF-κB expression. CONCLUSIONS Melatonin can inhibit the hyperactivity of NLRP3 inflammasomes by enhancing mitochondrial autophagy and inhibiting TLR4/NF-κB pathway activity. Thus, melatonin may be a promising treatment for alleviating the WMD caused by endotoxin and ischemic hypoxia in neonatal rats.
Collapse
Affiliation(s)
- Miao Qin
- Department of Neonatology, Affiliated Hospital of Qingdao University, Qingdao, China
| | | | | | | | | | | | | |
Collapse
|
25
|
Huang J, Yang J, Zou X, Zuo S, Wang J, Cheng J, Zhu H, Li W, Shi M, Zhao G, Liu Z. Ginkgolide B promotes oligodendrocyte precursor cell differentiation and survival via Akt/CREB/bcl-2 signaling pathway after white matter lesion. Exp Biol Med (Maywood) 2021; 246:1198-1209. [PMID: 33557607 PMCID: PMC8142115 DOI: 10.1177/1535370221989955] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 01/03/2021] [Indexed: 12/12/2022] Open
Abstract
White matter lesion (WML) is caused by chronic cerebral hypoperfusion, which are usually associated with cognitive impairment. Evidence from recent studies has shown that ginkgolide B has a neuroprotective effect that could be beneficial for the treatment of ischemia; however, it is not clear whether ginkgolide B has a protective effect on WML. Our data show that ginkgolide B can promote the differentiation of oligodendrocyte precursor cell (OPC) into oligodendrocytes and promote oligodendrocyte survival following a WML. Ginkgolide B (5, 10, 20 mg/kg) or saline is administered intraperitoneally every day after WML. After 4 weeks, the data of Morris water maze suggested that rats' memory and learning abilities were impaired, and the administration of ginkgolide B enhanced behavioral achievement. Also, treatment with ginkgolide B significantly attenuated this loss of myelin. Our result suggests that ginkgolide B promotes the differentiation of OPC into oligodendrocytes. We also found that ginkgolide B ameliorates oligodendrocytes apoptosis. Furthermore, ginkgolide B enhanced the expression of phosphorylated Akt and CREB. In conclusion, our data firstly show that ginkgolide B promotes oligodendrocyte genesis and oligodendrocyte myelin following a WML, possibly involving the Akt and CREB pathways.
Collapse
Affiliation(s)
- Jian Huang
- Department of Neurology, Xijing Hospital, Airforce Military Medical University, Xi'an, Shaanxi 710032, China
| | - Jun Yang
- Department of Nephrology, Xijing Hospital, Airforce Military Medical University, Xi'an, Shaanxi 710032, China
| | - Xingju Zou
- Department of Neurology, Xijing Hospital, Airforce Military Medical University, Xi'an, Shaanxi 710032, China
| | - Shilun Zuo
- Department of Neurology, Second Affiliated Hospital of Army Military Medical University, Chongqing 400038, China
| | - Jing Wang
- Department of Neurology, Xijing Hospital, Airforce Military Medical University, Xi'an, Shaanxi 710032, China
| | - Jing Cheng
- Department of Neurology, Xijing Hospital, Airforce Military Medical University, Xi'an, Shaanxi 710032, China
| | - Hao Zhu
- Department of Neurology, Xijing Hospital, Airforce Military Medical University, Xi'an, Shaanxi 710032, China
| | - Weiwang Li
- Department of Neurology, Xijing Hospital, Airforce Military Medical University, Xi'an, Shaanxi 710032, China
| | - Ming Shi
- Department of Neurology, Xijing Hospital, Airforce Military Medical University, Xi'an, Shaanxi 710032, China
| | - Gang Zhao
- Department of Neurology, Xijing Hospital, Airforce Military Medical University, Xi'an, Shaanxi 710032, China
| | - Zhirong Liu
- Department of Neurology, Xijing Hospital, Airforce Military Medical University, Xi'an, Shaanxi 710032, China
| |
Collapse
|
26
|
Aryal R, Patabendige A. Blood-brain barrier disruption in atrial fibrillation: a potential contributor to the increased risk of dementia and worsening of stroke outcomes? Open Biol 2021; 11:200396. [PMID: 33878948 PMCID: PMC8059575 DOI: 10.1098/rsob.200396] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Atrial fibrillation (AF) has become one of the most significant health problems worldwide, warranting urgent answers to currently pending questions on the effects of AF on brain function. Recent evidence has emerged to show an association between AF and an increased risk of developing dementia and worsening of stroke outcomes. A healthy brain is protected by the blood–brain barrier (BBB), which is formed by the endothelial cells that line cerebral capillaries. These endothelial cells are continuously exposed to shear stress (the frictional force generated by blood flow), which affects endothelial cell structure and function. Flow disturbances as experienced during AF can disrupt the BBB and leave the brain vulnerable to damage. Investigating the plausible mechanisms in detail, linking AF to cerebrovascular damage is difficult in humans, leading to paucity of available clinical data. Here, we discuss the available evidence for BBB disruption during AF due to altered cerebral blood flow, and how this may contribute to an increased risk of dementia and worsening of stroke outcomes.
Collapse
Affiliation(s)
- Ritambhara Aryal
- Brain Barriers Group, School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW 2308, Australia.,Brain and Mental Health Research Programme, Hunter Medical Research Institute, Newcastle, Australia
| | - Adjanie Patabendige
- Brain Barriers Group, School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW 2308, Australia.,Brain and Mental Health Research Programme, Hunter Medical Research Institute, Newcastle, Australia.,Institute of Infection and Global Health, University of Liverpool, Liverpool, UK
| |
Collapse
|
27
|
García-Bermúdez MY, Freude KK, Mouhammad ZA, van Wijngaarden P, Martin KK, Kolko M. Glial Cells in Glaucoma: Friends, Foes, and Potential Therapeutic Targets. Front Neurol 2021; 12:624983. [PMID: 33796062 PMCID: PMC8007906 DOI: 10.3389/fneur.2021.624983] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 01/26/2021] [Indexed: 12/15/2022] Open
Abstract
Glaucoma is the second leading cause of blindness worldwide, affecting ~80 million people by 2020 (1, 2). The condition is characterized by a progressive loss of retinal ganglion cells (RGCs) and their axons accompanied by visual field loss. The underlying pathophysiology of glaucoma remains elusive. Glaucoma is recognized as a multifactorial disease, and lowering intraocular pressure (IOP) is the only treatment that has been shown to slow the progression of the condition. However, a significant number of glaucoma patients continue to go blind despite intraocular pressure-lowering treatment (2). Thus, the need for alternative treatment strategies is indisputable. Accumulating evidence suggests that glial cells play a significant role in supporting RGC function and that glial dysfunction may contribute to optic nerve disease. Here, we review recent advances in understanding the role of glial cells in the pathophysiology of glaucoma. A particular focus is on the dynamic and essential interactions between glial cells and RGCs and potential therapeutic approaches to glaucoma by targeting glial cells.
Collapse
Affiliation(s)
| | - Kristine K Freude
- Department for Veterinary and Animal Science, University of Copenhagen, Copenhagen, Denmark
| | - Zaynab A Mouhammad
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Peter van Wijngaarden
- Center for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC, Australia.,Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, VIC, Australia
| | - Keith K Martin
- Center for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC, Australia.,Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, VIC, Australia
| | - Miriam Kolko
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark.,Department of Ophthalmology, Copenhagen University Hospital, Rigshospitalet-Glostrup, Glostrup, Denmark
| |
Collapse
|
28
|
Ping S, Qiu X, Kyle M, Zhao LR. Brain-derived CCR5 Contributes to Neuroprotection and Brain Repair after Experimental Stroke. Aging Dis 2021; 12:72-92. [PMID: 33532129 PMCID: PMC7801286 DOI: 10.14336/ad.2020.0406] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 04/06/2020] [Indexed: 02/04/2023] Open
Abstract
Chemokine (C-C motif) receptor 5 (CCR5) is expressed not only in the immune cells but also in cerebral cells such as neurons, glia, and vascular cells. Stroke triggers high expression of CCR5 in the brain. However, the role of CCR5 in stroke remains unclear. In this study, using bone marrow chimeras we have determined the involvement of brain-derived or bone marrow-derived CCR5 in neuroprotection and brain repair after experimental stroke. CCR5-/- mice that received either wild-type (WT) or CCR5-/- bone marrow transplantation showed larger infarction sizes than the WT mice that received either WT or CCR5-/- bone marrow transplantation in both the acute (48h) and subacute (2 months) phases after cerebral cortical ischemia, suggesting that the lack of CCR5 in the brain leads to severe brain damage after stroke. However, the lack of CCR5 in the bone marrow-derived cells did not affect infarction size. The impairments of somatosensory-motor function and motor coordination were exacerbated in the mice lacking CCR5 in the brain. At 2 months post-stroke, increased degenerative neurons, decreased dendrites and synapses, decreased Iba1+ microglia/ macrophages, reduced myelination and CNPase+ oligodendrocytes in the peri-infarct cortex were observed in the mice lacking CCR5 in the brain. These pathological changes are significantly correlated with the increased infarction size and exacerbated neurological deficits. These data suggest that brain-derived CCR5 plays a key role in neuroprotection and brain repair in the subacute phase of stroke. This study reveals a novel role of CCR5 in stroke, which sheds new light on post-stroke pathomechanism.
Collapse
Affiliation(s)
- Suning Ping
- Department of Neurosurgery, State University of New York Upstate Medical University, New York, USA
| | - Xuecheng Qiu
- Department of Neurosurgery, State University of New York Upstate Medical University, New York, USA
| | - Michele Kyle
- Department of Neurosurgery, State University of New York Upstate Medical University, New York, USA
| | - Li-Ru Zhao
- Department of Neurosurgery, State University of New York Upstate Medical University, New York, USA
| |
Collapse
|
29
|
Cui Y, Jin X, Choi JY, Kim BG. Modeling subcortical ischemic white matter injury in rodents: unmet need for a breakthrough in translational research. Neural Regen Res 2021; 16:638-642. [PMID: 33063714 PMCID: PMC8067929 DOI: 10.4103/1673-5374.295313] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Subcortical ischemic white matter injury (SIWMI), pathological correlate of white matter hyperintensities or leukoaraiosis on magnetic resonance imaging, is a common cause of cognitive decline in elderly. Despite its high prevalence, it remains unknown how various components of the white matter degenerate in response to chronic ischemia.This incomplete knowledge is in part due to a lack of adequate animal model. The current review introduces various SIWMI animal models and aims to scrutinize their advantages and disadvantages primarily in regard to the pathological manifestations of white matter components. The SIWMI animal models are categorized into 1) chemically induced SIWMI models, 2) vascular occlusive SIWMI models, and 3) SIWMI models with comorbid vascular risk factors. Chemically induced models display consistent lesions in predetermined areas of the white matter, but the abrupt evolution of lesions does not appropriately reflect the progressive pathological processes in human white matter hyperintensities. Vascular occlusive SIWMI models often do not exhibit white matter lesions that are sufficiently unequivocal to be quantified. When combined with comorbid vascular risk factors (specifically hypertension), however, they can produce progressive and definitive white matter lesions including diffuse rarefaction, demyelination, loss of oligodendrocytes, and glial activation, which are by far the closest to those found in human white matter hyperintensities lesions. However, considerable surgical mortality and unpredictable natural deaths during a follow-up period would necessitate further refinements in these models. In the meantime, in vitro SIWMI models that recapitulate myelinated white matter track may be utilized to study molecular mechanisms of the ischemic white matter injury. Appropriate in vivo and in vitro SIWMI models will contribute in a complementary manner to making a breakthrough in developing effective treatment to prevent progression of white matter hyperintensities.
Collapse
Affiliation(s)
- Yuexian Cui
- Department of Brain Science, Ajou University School of Medicine; Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Korea; Department of Neurology, Yanbian University Hospital, Yanji, Jilin Province, China
| | - Xuelian Jin
- Department of Brain Science, Ajou University School of Medicine; Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Korea; Department of Nephrology, Suqian First Hospital, Suqian, Jiangsu Province, China
| | - Jun Young Choi
- Department of Brain Science, Ajou University School of Medicine; Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University Graduate School of Medicine; Department of Neurology, Ajou University School of Medicine, Suwon, Korea
| | - Byung Gon Kim
- Department of Brain Science, Ajou University School of Medicine; Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University Graduate School of Medicine; Department of Neurology, Ajou University School of Medicine, Suwon, Korea
| |
Collapse
|
30
|
Quick S, Moss J, Rajani RM, Williams A. A Vessel for Change: Endothelial Dysfunction in Cerebral Small Vessel Disease. Trends Neurosci 2020; 44:289-305. [PMID: 33308877 DOI: 10.1016/j.tins.2020.11.003] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 09/24/2020] [Accepted: 11/11/2020] [Indexed: 01/08/2023]
Abstract
The blood vessels of the brain are lined with endothelial cells and it has been long known that these help to regulate blood flow to the brain. However, there is increasing evidence that these cells also interact with the surrounding brain tissue. These interactions change when the endothelial cells become dysfunctional and have an impact in diseases such as cerebral small vessel disease, the leading cause of vascular dementia. In this review, we focus on what endothelial dysfunction is, what causes it, how it leads to surrounding brain pathology, how researchers can investigate it with current models, and where this might lead in the future for dementia therapies.
Collapse
Affiliation(s)
- Sophie Quick
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Jonathan Moss
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Rikesh M Rajani
- UK Dementia Research Institute at UCL, University College London, London, UK
| | - Anna Williams
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK.
| |
Collapse
|
31
|
Shabir O, Moll TA, Matuszyk MM, Eyre B, Dake MD, Berwick J, Francis SE. Preclinical models of disease and multimorbidity with focus upon cardiovascular disease and dementia. Mech Ageing Dev 2020; 192:111361. [DOI: 10.1016/j.mad.2020.111361] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 08/28/2020] [Accepted: 09/16/2020] [Indexed: 12/12/2022]
|
32
|
Winder NR, Reeve EH, Walker AE. Large artery stiffness and brain health: insights from animal models. Am J Physiol Heart Circ Physiol 2020; 320:H424-H431. [PMID: 33164578 DOI: 10.1152/ajpheart.00696.2020] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
There are no effective treatments available to halt or reverse the progression of age-related cognitive decline and Alzheimer's disease. Thus, there is an urgent need to understand the underlying mechanisms of disease etiology and progression to identify novel therapeutic targets. Age-related changes to the vasculature, particularly increases in stiffness of the large elastic arteries, are now recognized as important contributors to brain aging. There is a growing body of evidence for an association between greater large artery stiffness and cognitive impairment among both healthy older adults and patients with Alzheimer's disease. However, studies in humans are limited to only correlative evidence, whereas animal models allow researchers to explore the causative mechanisms linking arterial stiffness to neurocognitive dysfunction and disease. Recently, several rodent models of direct modulation of large artery stiffness and the consequent effects on the brain have been reported. Common outcomes among these models have emerged, including evidence that greater large artery stiffness causes cerebrovascular dysfunction associated with increased oxidative stress and inflammatory signaling. The purpose of this mini-review is to highlight the recent findings associating large artery stiffness with deleterious brain outcomes, with a specific focus on causative evidence obtained from animal models. We will also discuss the gaps in knowledge that remain in our understanding of how large artery stiffness affects brain function and disease outcomes.
Collapse
Affiliation(s)
- Nick R Winder
- Department of Human Physiology, University of Oregon, Eugene, Oregon
| | - Emily H Reeve
- Department of Human Physiology, University of Oregon, Eugene, Oregon
| | - Ashley E Walker
- Department of Human Physiology, University of Oregon, Eugene, Oregon
| |
Collapse
|
33
|
Tuo QZ, Zou JJ, Lei P. Rodent Models of Vascular Cognitive Impairment. J Mol Neurosci 2020; 71:1-12. [DOI: 10.1007/s12031-020-01733-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 10/12/2020] [Indexed: 12/23/2022]
|
34
|
Zhang L, Ellor S, Sun JM, Liu C, Kurtzburg J, Song AW. DTI Tract-Based Quantitative Susceptibility Mapping: An Initial Feasibility Study to Investigate the Potential Role of Myelination in Brain Connectivity Change in Cerebral Palsy Patients During Autologous Cord Blood Cell Therapy Using a Rotationally-Invariant Quantitative Measure. J Magn Reson Imaging 2020; 53:251-258. [PMID: 32677156 DOI: 10.1002/jmri.27286] [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: 04/07/2020] [Revised: 06/24/2020] [Accepted: 06/26/2020] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Previous studies using diffusion tensor imaging (DTI)-based connectome analysis revealed improved connectivity in cerebral palsy (CP) patients who underwent autologous umbilical cord blood (UCB) stem-cell therapy. However, the potential mechanism for the connectivity increase remains unclear and needs to be further elucidated. PURPOSE To develop a technique with improved accuracy for quantitative susceptibility mapping (QSM) with unique sensitivity to myelin, and demonstrate its use in elucidating the underlying mechanism of the observed motor function improvement and brain connectivity increase in CP patients who received autologous UCB stem-cell therapy. STUDY TYPE Prospective. POPULATION A cohort of eight pediatric CP patients (2.6 ± 0.6 years of age) with intact corticospinal tracts (CST) from a randomized, placebo-controlled trial of autologous UCB stem-cell therapy in CP children was included in this study. FIELD STRENGTH/SEQUENCE DTI and 3D spoiled gradient recalled (SPGR) QSM at 3.0T. ASSESSMENT Pre- and posttreatment magnetic susceptibility (χ) and the rotationally-invariant magnetic susceptibility anisotropy (MSA) along the CST were derived. Behavioral changes were assessed using the 66-item Gross Motor Function Measurement. Changes in χ and MSA were compared between patients with and without substantial behavioral improvements. STATISTICAL TESTS Two-sample t-tests were performed to assess the differences in the changes of measurements of interest (Δχ, ΔMSA, and ΔFA) between patients who significantly improved and those who did not. RESULTS Patients who demonstrated posttreatment motor improvements exceeding expectations showed significantly more diamagnetic Δχ in the periventricular region along the CST (P = 0.003). Further analysis on the ΔMSA of this region was significantly increased (P = 0.006) for high responders, along with concurrent FA increase. DATA CONCLUSION These initial findings suggest that the DTI tract-based QSM method has the potential to characterize white matter changes associated with behavioral improvements in CP children who underwent cord blood stem-cell therapy. LEVEL OF EVIDENCE 2 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Lijia Zhang
- Brain Imaging and Analysis Center, Duke University Medical Center, Duke University, Durham, North Carolina, USA
| | - Susan Ellor
- Department of Neurology, Johns Hopkins Bayview Medical Center, Baltimore, Maryland, USA
| | - Jessica M Sun
- The Robertson Clinical and Translational Cell Therapy Program, Duke University, Durham, North California, USA
| | - Chunlei Liu
- Department of Electrical Engineering and Computer Sciences, University of California Berkeley, Berkeley, California, USA.,Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, California, USA
| | - Joanne Kurtzburg
- The Robertson Clinical and Translational Cell Therapy Program, Duke University, Durham, North California, USA
| | - Allen W Song
- Brain Imaging and Analysis Center, Duke University Medical Center, Duke University, Durham, North Carolina, USA
| |
Collapse
|
35
|
Lefevere E, Salinas‐Navarro M, Andries L, Noterdaeme L, Etienne I, Van Wonterghem E, Vinckier S, Davis BM, Van Bergen T, Van Hove I, Movahedi K, Vandenbroucke RE, Moons L, De Groef L. Tightening the retinal glia limitans attenuates neuroinflammation after optic nerve injury. Glia 2020; 68:2643-2660. [DOI: 10.1002/glia.23875] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 05/20/2020] [Accepted: 06/04/2020] [Indexed: 12/14/2022]
Affiliation(s)
- Evy Lefevere
- Neural Circuit Development and Regeneration Research Group, Department of Biology KU Leuven Leuven Belgium
- Leuven Brain Institute (LBI) KU Leuven Leuven Belgium
| | - Manuel Salinas‐Navarro
- Neural Circuit Development and Regeneration Research Group, Department of Biology KU Leuven Leuven Belgium
- Leuven Brain Institute (LBI) KU Leuven Leuven Belgium
| | - Lien Andries
- Neural Circuit Development and Regeneration Research Group, Department of Biology KU Leuven Leuven Belgium
- Leuven Brain Institute (LBI) KU Leuven Leuven Belgium
| | - Lut Noterdaeme
- Neural Circuit Development and Regeneration Research Group, Department of Biology KU Leuven Leuven Belgium
| | | | - Elien Van Wonterghem
- Barriers in Inflammation Lab VIB Center for Inflammation Research Ghent Belgium
- Department of Biomedical Molecular Biology Ghent University Ghent Belgium
| | - Stefan Vinckier
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, and Department of Oncology and Leuven Cancer Institute (LKI) VIB and KU Leuven Leuven Belgium
| | - Benjamin M. Davis
- Glaucoma and Retinal Neurodegeneration Research, Visual Neuroscience UCL Institute of Ophthalmology London UK
| | | | - Inge Van Hove
- Neural Circuit Development and Regeneration Research Group, Department of Biology KU Leuven Leuven Belgium
- Oxurion NV Leuven Belgium
| | - Kiavash Movahedi
- Myeloid Cell Immunology Lab VIB Center for Inflammation Research Brussels Belgium
- Lab of Cellular and Molecular Immunology Vrije Universiteit Brussel Brussels Belgium
| | - Roosmarijn E. Vandenbroucke
- Barriers in Inflammation Lab VIB Center for Inflammation Research Ghent Belgium
- Department of Biomedical Molecular Biology Ghent University Ghent Belgium
| | - Lieve Moons
- Neural Circuit Development and Regeneration Research Group, Department of Biology KU Leuven Leuven Belgium
- Leuven Brain Institute (LBI) KU Leuven Leuven Belgium
| | - Lies De Groef
- Neural Circuit Development and Regeneration Research Group, Department of Biology KU Leuven Leuven Belgium
- Leuven Brain Institute (LBI) KU Leuven Leuven Belgium
| |
Collapse
|
36
|
Manukjan N, Ahmed Z, Fulton D, Blankesteijn WM, Foulquier S. A Systematic Review of WNT Signaling in Endothelial Cell Oligodendrocyte Interactions: Potential Relevance to Cerebral Small Vessel Disease. Cells 2020; 9:cells9061545. [PMID: 32630426 PMCID: PMC7349551 DOI: 10.3390/cells9061545] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 06/19/2020] [Accepted: 06/23/2020] [Indexed: 12/21/2022] Open
Abstract
Key pathological features of cerebral small vessel disease (cSVD) include impairment of the blood brain barrier (BBB) and the progression of white matter lesions (WMLs) amongst other structural lesions, leading to the clinical manifestations of cSVD. The function of endothelial cells (ECs) is of major importance to maintain a proper BBB. ECs interact with several cell types to provide structural and functional support to the brain. Oligodendrocytes (OLs) myelinate axons in the central nervous system and are crucial in sustaining the integrity of white matter. The interplay between ECs and OLs and their precursor cells (OPCs) has received limited attention yet seems of relevance for the study of BBB dysfunction and white matter injury in cSVD. Emerging evidence shows a crosstalk between ECs and OPCs/OLs, mediated by signaling through the Wingless and Int-1 (WNT)/β-catenin pathway. As the latter is involved in EC function (e.g., angiogenesis) and oligodendrogenesis, we reviewed the role of WNT/β-catenin signaling for both cell types and performed a systematic search to identify studies describing a WNT-mediated interplay between ECs and OPCs/OLs. Dysregulation of this interaction may limit remyelination of WMLs and render the BBB leaky, thereby initiating a vicious neuroinflammatory cycle. A better understanding of the role of this signaling pathway in EC-OL crosstalk is essential in understanding cSVD development.
Collapse
Affiliation(s)
- Narek Manukjan
- Department of Pharmacology and Toxicology, Maastricht University, PO Box 616, 6200 MD Maastricht, The Netherlands; or (W.M.B.)
- CARIM—School for Cardiovascular Diseases, Maastricht University Medical Center+, PO Box 616, 6200 MD Maastricht, The Netherlands
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK; (Z.A.); (D.F.)
| | - Zubair Ahmed
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK; (Z.A.); (D.F.)
| | - Daniel Fulton
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK; (Z.A.); (D.F.)
| | - W. Matthijs Blankesteijn
- Department of Pharmacology and Toxicology, Maastricht University, PO Box 616, 6200 MD Maastricht, The Netherlands; or (W.M.B.)
- CARIM—School for Cardiovascular Diseases, Maastricht University Medical Center+, PO Box 616, 6200 MD Maastricht, The Netherlands
| | - Sébastien Foulquier
- Department of Pharmacology and Toxicology, Maastricht University, PO Box 616, 6200 MD Maastricht, The Netherlands; or (W.M.B.)
- CARIM—School for Cardiovascular Diseases, Maastricht University Medical Center+, PO Box 616, 6200 MD Maastricht, The Netherlands
- Department of Neurology, MHeNs—School for Mental Health and Neuroscience, Maastricht University Medical Center+, PO Box 616, 6200 MD Maastricht, The Netherlands
- Correspondence: ; Tel.: +31-43-3881409
| |
Collapse
|
37
|
Liang YQ, Kakino A, Matsuzaka Y, Mashimo T, Isono M, Akamatsu T, Shimizu H, Tajima M, Kaneko T, Li L, Takeuchi F, Sawamura T, Kato N. LOX-1 (Lectin-Like Oxidized Low-Density Lipoprotein Receptor-1) Deletion Has Protective Effects on Stroke in the Genetic Background of Stroke-Prone Spontaneously Hypertensive Rat. Stroke 2020; 51:1835-1843. [PMID: 32397936 DOI: 10.1161/strokeaha.120.029421] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Background and Purpose- oxLDL (oxidized low-density lipoprotein) has been known for its potential to induce endothelial dysfunction and used as a major serological marker of oxidative stress. Recently, LOX-1 (lectin-like oxidized low-density lipoprotein receptor-1), a lectin-like receptor for oxLDL, has attracted attention in studies of neuronal apoptosis and stroke. We aim to investigate the impact of LOX-1-deficiency on spontaneous hypertension-related brain damage in the present study. Methods- We generated a LOX-1 deficient strain on the genetic background of stroke-prone spontaneously hypertensive rat (SHRSP), an animal model of severe hypertension and spontaneous stroke. In this new disease model with stroke-proneness, we monitored the occurrence of brain abnormalities with and without salt loading by multiple procedures including T2 weighted magnetic resonance imaging and also explored circulatory miRNAs as diagnostic biomarkers for cerebral ischemic injury by microarray analysis. Results- Both T2 weighted magnetic resonance imaging abnormalities and physiological parameter changes could be detected at significantly delayed timing in LOX-1 knockout rats compared with wild-type SHRSP, in either case of normal rat chow and salt loading (P<0.005 in all instances; n=11-20 for SHRSP and n=13-23 for LOX-1 knockout rats). There were no significant differences in the form of magnetic resonance imaging findings between the strains. A number of miRNAs expressed in the normal rat plasma, including rno-miR-150-5p and rno-miR-320-3p, showed significant changes after spontaneous brain damage in SHRSP, whereas the corresponding changes were modest or almost unnoticeable in LOX-1 knockout rats. There appeared to be the lessening of correlation of postischemic miRNA alterations between the injured brain tissue and plasma in LOX-1 knockout rats. Conclusions- Our data show that deficiency of LOX-1 has a protective effect on spontaneous brain damage in a newly generated LOX-1-deficient strain of SHRSP. Further, our analysis of miRNAs as biomarkers for ischemic brain damage supports a potential involvement of LOX-1 in blood brain barrier disruption after cerebral ischemia. Visual Overview- An online visual overview is available for this article.
Collapse
Affiliation(s)
- Yi-Qiang Liang
- From the Department of Gene Diagnostics and Therapeutics (Y.-Q.L., Y.M., M.I., T.A., H.S., M.T., F.T., N.K.), National Center for Global Health and Medicine
| | - Akemi Kakino
- Department of Molecular Pathophysiology, Shinshu University School of Medicine (A.K., T.S.).,Institute for Biomedical Sciences, Shinshu University (A.K., T.S.)
| | - Yasunari Matsuzaka
- From the Department of Gene Diagnostics and Therapeutics (Y.-Q.L., Y.M., M.I., T.A., H.S., M.T., F.T., N.K.), National Center for Global Health and Medicine
| | - Tomoji Mashimo
- Institute of Medical Science, The University of Tokyo (T.M.).,Institute of Experimental Animal Sciences Graduate School of Medicine, Osaka University (T.M.)
| | - Masato Isono
- From the Department of Gene Diagnostics and Therapeutics (Y.-Q.L., Y.M., M.I., T.A., H.S., M.T., F.T., N.K.), National Center for Global Health and Medicine
| | - Tomohisa Akamatsu
- From the Department of Gene Diagnostics and Therapeutics (Y.-Q.L., Y.M., M.I., T.A., H.S., M.T., F.T., N.K.), National Center for Global Health and Medicine.,Department of Pediatrics (T.A.), National Center for Global Health and Medicine
| | - Hana Shimizu
- From the Department of Gene Diagnostics and Therapeutics (Y.-Q.L., Y.M., M.I., T.A., H.S., M.T., F.T., N.K.), National Center for Global Health and Medicine
| | - Michiko Tajima
- From the Department of Gene Diagnostics and Therapeutics (Y.-Q.L., Y.M., M.I., T.A., H.S., M.T., F.T., N.K.), National Center for Global Health and Medicine
| | - Takehito Kaneko
- Graduate School of Science and Engineering, Iwate University (T.K.)
| | - Lei Li
- Department of Vascular Physiology, National Cerebral and Cardiovascular Center (L.L.)
| | - Fumihiko Takeuchi
- From the Department of Gene Diagnostics and Therapeutics (Y.-Q.L., Y.M., M.I., T.A., H.S., M.T., F.T., N.K.), National Center for Global Health and Medicine
| | - Tatsuya Sawamura
- Department of Molecular Pathophysiology, Shinshu University School of Medicine (A.K., T.S.).,Institute for Biomedical Sciences, Shinshu University (A.K., T.S.)
| | - Norihiro Kato
- From the Department of Gene Diagnostics and Therapeutics (Y.-Q.L., Y.M., M.I., T.A., H.S., M.T., F.T., N.K.), National Center for Global Health and Medicine
| |
Collapse
|
38
|
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: 332] [Impact Index Per Article: 83.0] [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
|
39
|
Gong X, Shan W, Yuan K, Lu Z, Zhang M, Lu J, Zhang X, Huang X, Guo H, Peng M, Liu X, Zhao X, Xu G. Dietary Inflammatory Index and Leukoaraiosis in Patients with Ischemic Stroke. J Nutr Health Aging 2020; 24:473-477. [PMID: 32346684 DOI: 10.1007/s12603-020-1351-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Diet may change the chronic levels of systemic inflammation, which in turn influence the development of leukoaraiosis (LA). This study aimed to examine the association between dietary inflammatory index (DII) and LA in patients with ischemic stroke. METHODS Patients with first-ever ischemic stroke were enrolled from two centers. A semi-quantitative food frequency questionnaire (FFQ) was used to evaluate diet contents. The DII score of each patient was calculated based on the reported diet contents. Presence and degree of LA were evaluated with a magnetic resonance imaging (MRI) scan. LA was graded according to Fazekas scale. RESULTS Of the 497 enrolled patients, 337 (67.8%) were detected with LA. Patients with LA had a higher DII score (0.23 vs -0.88, P < 0.001). Logistic regression analysis detected that patients with highest quartile of DII score had an OR of 3.61 (95% CI: 2.05-6.36, P < 0.001) for LA compared with those with lowest quartile of DII. After adjusting for major confounders, the highest DII quartile remained as an independent predictor for LA (OR = 2.66, 95% CI: 1.41-5.00, P = 0.008). CONCLUSIONS A pro-inflammatory diet pattern, as indicated by higher DII values, appears to be associated with a higher risk of LA. This result suggested that dietary-mediated inflammation may involved in the pathogenesis of LA, which warrant further study.
Collapse
Affiliation(s)
- X Gong
- Gelin Xu, Department of Neurology, Jinling Hospital, First School of Clinical Medicine, Southern Medical University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu Province, China. Tel: (+) 86- 18951919349; E-Mail: ; Xiongfei Zhao, Department of Neurology, Cardiovascular and Cerebrovascular Disease Hospital of Meishan, Meishan 620000, Sichuan, China. Tel: (+) 86-13609147368;
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Lerman LO, Kurtz TW, Touyz RM, Ellison DH, Chade AR, Crowley SD, Mattson DL, Mullins JJ, Osborn J, Eirin A, Reckelhoff JF, Iadecola C, Coffman TM. Animal Models of Hypertension: A Scientific Statement From the American Heart Association. Hypertension 2019; 73:e87-e120. [PMID: 30866654 DOI: 10.1161/hyp.0000000000000090] [Citation(s) in RCA: 171] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Hypertension is the most common chronic disease in the world, yet the precise cause of elevated blood pressure often cannot be determined. Animal models have been useful for unraveling the pathogenesis of hypertension and for testing novel therapeutic strategies. The utility of animal models for improving the understanding of the pathogenesis, prevention, and treatment of hypertension and its comorbidities depends on their validity for representing human forms of hypertension, including responses to therapy, and on the quality of studies in those models (such as reproducibility and experimental design). Important unmet needs in this field include the development of models that mimic the discrete hypertensive syndromes that now populate the clinic, resolution of ongoing controversies in the pathogenesis of hypertension, and the development of new avenues for preventing and treating hypertension and its complications. Animal models may indeed be useful for addressing these unmet needs.
Collapse
|
41
|
Denver P, D’Adamo H, Hu S, Zuo X, Zhu C, Okuma C, Kim P, Castro D, Jones MR, Leal C, Mekkittikul M, Ghadishah E, Teter B, Vinters HV, Cole GM, Frautschy SA. A Novel Model of Mixed Vascular Dementia Incorporating Hypertension in a Rat Model of Alzheimer's Disease. Front Physiol 2019; 10:1269. [PMID: 31708792 PMCID: PMC6821690 DOI: 10.3389/fphys.2019.01269] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 09/19/2019] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD) and mixed dementia (MxD) comprise the majority of dementia cases in the growing global aging population. MxD describes the coexistence of AD pathology with vascular pathology, including cerebral small vessel disease (SVD). Cardiovascular disease increases risk for AD and MxD, but mechanistic synergisms between the coexisting pathologies affecting dementia risk, progression and the ultimate clinical manifestations remain elusive. To explore the additive or synergistic interactions between AD and chronic hypertension, we developed a rat model of MxD, produced by breeding APPswe/PS1ΔE9 transgenes into the stroke-prone spontaneously hypertensive rat (SHRSP) background, resulting in the SHRSP/FAD model and three control groups (FAD, SHRSP and non-hypertensive WKY rats, n = 8-11, both sexes, 16-18 months of age). After behavioral testing, rats were euthanized, and tissue assessed for vascular, neuroinflammatory and AD pathology. Hypertension was preserved in the SHRSP/FAD cross. Results showed that SHRSP increased FAD-dependent neuroinflammation (microglia and astrocytes) and tau pathology, but plaque pathology changes were subtle, including fewer plaques with compact cores and slightly reduced plaque burden. Evidence for vascular pathology included a change in the distribution of astrocytic end-foot protein aquaporin-4, normally distributed in microvessels, but in SHRSP/FAD rats largely dissociated from vessels, appearing disorganized or redistributed into neuropil. Other evidence of SVD-like pathology included increased collagen IV staining in cerebral vessels and PECAM1 levels. We identified a plasma biomarker in SHRSP/FAD rats that was the only group to show increased Aqp-4 in plasma exosomes. Evidence of neuron damage in SHRSP/FAD rats included increased caspase-cleaved actin, loss of myelin and reduced calbindin staining in neurons. Further, there were mitochondrial deficits specific to SHRSP/FAD, notably the loss of complex II, accompanying FAD-dependent loss of mitochondrial complex I. Cognitive deficits exhibited by FAD rats were not exacerbated by the introduction of the SHRSP phenotype, nor was the hyperactivity phenotype associated with SHRSP altered by the FAD transgene. This novel rat model of MxD, encompassing an amyloidogenic transgene with a hypertensive phenotype, exhibits several features associated with human vascular or "mixed" dementia and may be a useful tool in delineating the pathophysiology of MxD and development of therapeutics.
Collapse
Affiliation(s)
- Paul Denver
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Heather D’Adamo
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Shuxin Hu
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
- Geriatric Research Education and Clinical Center, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, CA, United States
| | - Xiaohong Zuo
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
- Geriatric Research Education and Clinical Center, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, CA, United States
| | - Cansheng Zhu
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - Chihiro Okuma
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - Peter Kim
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
- Geriatric Research Education and Clinical Center, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, CA, United States
| | - Daniel Castro
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - Mychica R. Jones
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - Carmen Leal
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
- Geriatric Research Education and Clinical Center, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, CA, United States
| | - Marisa Mekkittikul
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
- Geriatric Research Education and Clinical Center, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, CA, United States
| | - Elham Ghadishah
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Bruce Teter
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Geriatric Research Education and Clinical Center, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, CA, United States
| | - Harry V. Vinters
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - Gregory Michael Cole
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Geriatric Research Education and Clinical Center, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, CA, United States
| | - Sally A. Frautschy
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Geriatric Research Education and Clinical Center, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, CA, United States
| |
Collapse
|
42
|
Du B, Li H, Zheng H, Fan C, Liang M, Lian Y, Wei Z, Zhang Y, Bi X. Minocycline Ameliorates Depressive-Like Behavior and Demyelination Induced by Transient Global Cerebral Ischemia by Inhibiting Microglial Activation. Front Pharmacol 2019; 10:1247. [PMID: 31695615 PMCID: PMC6817504 DOI: 10.3389/fphar.2019.01247] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 09/27/2019] [Indexed: 12/19/2022] Open
Abstract
Global cerebral ischemia (GCI) commonly occurs in the elderly. Subcortical white matter lesions and oligodendrocyte (OLG) loss caused by cerebral ischemia have been implicated in the development of post-ischemic depression and cognitive impairment. OLGs are necessary for axonal myelination; the disrupted differentiation of OLG progenitor cells (OPCs) is associated with impaired remyelination. Evidence has indicated that increased levels of inflammatory cytokines released from activated microglia induce depression-like behaviors by affecting neurotransmitter pathways, but the mechanisms remain elusive. We explored the potential mechanisms that link microglia activation with GCI-induced depression and cognitive dysfunction by studying effects of minocycline on white matter damage, cytokine levels, and the monoaminergic neurotransmitters. An acute GCI animal model was generated through bilateral common carotid artery occlusion to induce ischemic inflammation and subcortical white matter damage. Minocycline, an inhibitor of microglia activation, was intraperitoneally administrated immediately after surgery and continued daily for additional six days. Minocycline shortened the immobile duration in tail suspension test and forced swimming test, while no improvement was found in Morris water maze test. The plasma levels of IL-1β, IL-6, TNF-α, HMGB1, and netrin-1 were significantly reduced with the treatment of minocycline. Minocycline treatment substantially reversed demyelination in corpus callosum and hippocampus, alleviated hippocampal microglia activation, and promoted OPCs maturation, while no effect was found on hippocampal neurodegeneration. Besides, the content of dopamine (DA) in the hippocampus was upregulated by minocycline treatment after GCI. Collectively, our data demonstrated that minocycline exerts an anti-depressant effect by inhibiting microglia activation, promoting OPCs maturation and remyelination. Increased DA in hippocampus may also play a role in ameliorating depressive behavior with minocycline treatment.
Collapse
Affiliation(s)
- Bingying Du
- Department of Neurology, Shanghai Changhai Hospital, the Second Military Medical University, Shanghai, China.,Department of Neurology, General Hospital of Central Theater Command of Chinese People's Liberation Army, Wuhan, China
| | - Hailong Li
- Department of Neurology, Shanghai Changhai Hospital, the Second Military Medical University, Shanghai, China.,Department of Rehabilitation Medicine, Zhejiang Hospital, Hangzhou, China
| | - Huiwen Zheng
- Department of Neurology, Shanghai Changhai Hospital, the Second Military Medical University, Shanghai, China
| | - Cunxiu Fan
- Department of Neurology, Shanghai Changhai Hospital, the Second Military Medical University, Shanghai, China
| | - Meng Liang
- Department of Neurology, Shanghai Changhai Hospital, the Second Military Medical University, Shanghai, China
| | - Yongjie Lian
- Department of Neurology, Shanghai Changhai Hospital, the Second Military Medical University, Shanghai, China
| | - Zelan Wei
- Department of Psychiatry, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Yanbo Zhang
- Department of Psychiatry, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Xiaoying Bi
- Department of Neurology, Shanghai Changhai Hospital, the Second Military Medical University, Shanghai, China
| |
Collapse
|
43
|
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: 2.0] [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
|
44
|
Low A, Mak E, Rowe JB, Markus HS, O'Brien JT. Inflammation and cerebral small vessel disease: A systematic review. Ageing Res Rev 2019; 53:100916. [PMID: 31181331 DOI: 10.1016/j.arr.2019.100916] [Citation(s) in RCA: 223] [Impact Index Per Article: 44.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 05/23/2019] [Accepted: 06/05/2019] [Indexed: 12/13/2022]
Abstract
Inflammation is increasingly implicated as a risk factor for dementia, stroke, and small vessel disease (SVD). However, the underlying mechanisms and causative pathways remain unclear. We systematically reviewed the existing literature on the associations between markers of inflammation and SVD (i.e., white matter hyperintensities (WMH), lacunes, enlarged perivascular spaces (EPVS), cerebral microbleeds (CMB)) in cohorts of older people with good health, cerebrovascular disease, or cognitive impairment. Based on distinctions made in the literature, markers of inflammation were classified as systemic inflammation (e.g. C-reactive protein, interleukin-6, fibrinogen) or vascular inflammation/endothelial dysfunction (e.g. homocysteine, von Willebrand factor, Lp-PLA2). Evidence from 82 articles revealed relatively robust associations between SVD and markers of vascular inflammation, especially amongst stroke patients, suggesting that alterations to the endothelium and blood-brain barrier may be a driving force behind SVD. Conversely, cross-sectional findings on systemic inflammation were mixed, although longitudinal investigations demonstrated that elevated levels of systemic inflammatory markers at baseline predicted subsequent SVD severity and progression. Importantly, regional analysis revealed that systemic and vascular inflammation were differentially related to two distinct forms of SVD. Specifically, markers of vascular inflammation tended to be associated with SVD in areas typical of hypertensive arteriopathy (e.g., basal ganglia), while systemic inflammation appeared to be involved in CAA-related vascular damage (e.g., centrum semiovale). Nonetheless, there is insufficient data to establish whether inflammation is causal of, or secondary to, SVD. Findings have important implications on interventions, suggesting the potential utility of treatments targeting the brain endothelium and blood brain barrier to combat SVD and associated neurodegenerative diseases.
Collapse
Affiliation(s)
- Audrey Low
- Department of Psychiatry, University of Cambridge, United Kingdom
| | - Elijah Mak
- Department of Psychiatry, University of Cambridge, United Kingdom
| | - James B Rowe
- Department of Clinical Neurosciences, University of Cambridge, United Kingdom
| | - Hugh S Markus
- Department of Clinical Neurosciences, University of Cambridge, United Kingdom
| | - John T O'Brien
- Department of Psychiatry, University of Cambridge, United Kingdom.
| |
Collapse
|
45
|
Brown RB, Traylor M, Burgess S, Sawcer S, Markus HS. Do Cerebral Small Vessel Disease and Multiple Sclerosis Share Common Mechanisms of White Matter Injury? Stroke 2019; 50:1968-1972. [PMID: 31221055 PMCID: PMC6661245 DOI: 10.1161/strokeaha.118.023649] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 04/18/2019] [Accepted: 05/01/2019] [Indexed: 12/21/2022]
Abstract
Background and Purpose- The role of inflammation in ischemic white matter disease is increasingly recognized, and further understanding of the pathophysiology might inform future treatment strategies. Multiple sclerosis (MS) is a chronic autoimmune condition in which inflammation plays a central role that also affects the white matter. We hypothesized that white matter injury might share common mechanisms and used statistical genetics techniques to assess whether having genetically elevated white matter hyperintensity (WMH) volume was associated with increased MS risk. Methods- We investigated the genetic association in 2 cohorts with magnetic resonance imaging-quantified ischemic white matter lesion volume (WMH in stroke; n=2797 and UK Biobank; n=8353) and 14 802 cases of MS and 26 703 controls from the International Multiple Sclerosis Genetics Consortium. We further performed individual-level polygenic risk score calculations for MS and measures of structural white matter disease in UK Biobank. Finally, we looked for evidence of overlapping risk across the whole genome. Results- There was no association of genetic variants influencing MS with WMH volume using summary statistics in the WMH in stroke cohort (relative risk score =1.014; 95% CI, 0.936-1.110) or in the UK Biobank cohort (relative risk score =1.030; 95% CI, 0.932-1.117). Conversely, assessing the contribution of single nucleotide polymorphisms significantly associated with WMH on the risk of MS there was no significant association (relative risk score =0.930; 95% CI, 0.736-1.191). There were no significant associations between polygenic risk scores calculations; these results were robust to the selection of single nucleotide polymorphisms at a range of significance thresholds. Whole genome analysis did not reveal any overlap of risk between the traits. Conclusions- Our results do not provide evidence to suggest a shared mechanism of white matter damage in ischemia and MS. We propose that inflammation acts in distinct pathways because of the differing nature of the primary insult.
Collapse
Affiliation(s)
- Robin B. Brown
- From the Department of Clinical Neurosciences (R.B.B., M.T., S.S., H.S.M.), University of Cambridge, United Kingdom
| | - Matthew Traylor
- From the Department of Clinical Neurosciences (R.B.B., M.T., S.S., H.S.M.), University of Cambridge, United Kingdom
| | - Stephen Burgess
- Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care (S.B.), University of Cambridge, United Kingdom
- MRC Biostatistics Unit, Cambridge Institute of Public Health, United Kingdom (S.B.)
| | - Stephen Sawcer
- From the Department of Clinical Neurosciences (R.B.B., M.T., S.S., H.S.M.), University of Cambridge, United Kingdom
| | - Hugh S. Markus
- From the Department of Clinical Neurosciences (R.B.B., M.T., S.S., H.S.M.), University of Cambridge, United Kingdom
| |
Collapse
|
46
|
Herisson F, Zhou I, Mawet J, Du E, Barfejani AH, Qin T, Cipolla MJ, Sun PZ, Rost NS, Ayata C. Posterior reversible encephalopathy syndrome in stroke-prone spontaneously hypertensive rats on high-salt diet. J Cereb Blood Flow Metab 2019; 39:1232-1246. [PMID: 29350576 PMCID: PMC6668522 DOI: 10.1177/0271678x17752795] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Stroke-prone spontaneously hypertensive rats (SHRSP) on high-salt diet are characterized by extremely high arterial pressures, and have been endorsed as a model for hypertensive small vessel disease and vascular cognitive impairment. However, rapidly developing malignant hypertension is a well-known cause of posterior reversible encephalopathy syndrome (PRES) in humans, associated with acute neurological deficits, seizures, vasogenic cerebral edema and microhemorrhages. In this study, we aimed to examine the overlap between human PRES and SHRSP on high-salt diet. In SHRSP, arterial blood pressure progressively increased after the onset of high-salt diet and seizure-like signs emerged within three to five weeks. MRI revealed progressive T2-hyperintense lesions suggestive of vasogenic edema predominantly in the cortical watershed and white matter regions. Histopathology confirmed severe blood-brain barrier disruption, white matter vacuolization and microbleeds that were more severe posteriorly. Hematological data suggested a thrombotic microangiopathy as a potential underlying mechanism. Unilateral common carotid artery occlusion protected the ipsilateral hemisphere from neuropathological abnormalities. Notably, all MRI and histopathological abnormalities were acutely reversible upon switching to regular diet and starting antihypertensive treatment. Altogether our data suggest that SHRSP on high-salt diet recapitulates the neurological, histopathological and imaging features of human PRES rather than chronic progressive small vessel disease.
Collapse
Affiliation(s)
- Fanny Herisson
- 1 Department of Radiology, Neurovascular Research Laboratory, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Iris Zhou
- 2 Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Jerome Mawet
- 1 Department of Radiology, Neurovascular Research Laboratory, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA.,3 Emergency Headache Center, Department of Neurology, Lariboisière Hospital, APHP, Sorbonne Paris-Cité, Paris, France
| | - E Du
- 4 Department of Ocean and Mechanical Engineering, Florida Atlantic University, Boca Raton, FL USA
| | - Arnavaz H Barfejani
- 1 Department of Radiology, Neurovascular Research Laboratory, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Tao Qin
- 1 Department of Radiology, Neurovascular Research Laboratory, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Marilyn J Cipolla
- 5 Department of Neurological Sciences, University of Vermont, Burlington, VT, USA
| | - Philip Z Sun
- 2 Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Natalia S Rost
- 6 J. Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Cenk Ayata
- 1 Department of Radiology, Neurovascular Research Laboratory, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA.,7 Department of Neurology, Stroke Service, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
47
|
Cipollini V, Troili F, Giubilei F. Emerging Biomarkers in Vascular Cognitive Impairment and Dementia: From Pathophysiological Pathways to Clinical Application. Int J Mol Sci 2019; 20:ijms20112812. [PMID: 31181792 PMCID: PMC6600494 DOI: 10.3390/ijms20112812] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 05/30/2019] [Accepted: 06/03/2019] [Indexed: 12/13/2022] Open
Abstract
Vascular pathology is the second most common neuropathology of dementia after Alzheimer’s disease (AD), with small vessels disease (SVD) being considered the major cause of vascular cognitive impairment and dementia (VCID). This review aims to evaluate pathophysiological pathways underlying a diagnosis of VCID. Firstly, we will discuss the role of endothelial dysfunction, blood-brain barrier disruption and neuroinflammation in its pathogenesis. Then, we will analyse different biomarkers including the ones of inflammatory responses to central nervous system tissue injuries, of coagulation and thrombosis and of circulating microRNA. Evidences on peripheral biomarkers for VCID are still poor and large-scale, prospectively designed studies are needed to translate these findings into clinical practice, in order to set different combinations of biomarkers to use for differential diagnosis among types of dementia.
Collapse
Affiliation(s)
- Virginia Cipollini
- S. Andrea Hospital, NESMOS Department, Faculty of Medicine and Psychology, Sapienza University of Rome, Via di Grottarossa 1035, 00189 Roma, Italy.
| | - Fernanda Troili
- S. Andrea Hospital, NESMOS Department, Faculty of Medicine and Psychology, Sapienza University of Rome, Via di Grottarossa 1035, 00189 Roma, Italy.
| | - Franco Giubilei
- S. Andrea Hospital, NESMOS Department, Faculty of Medicine and Psychology, Sapienza University of Rome, Via di Grottarossa 1035, 00189 Roma, Italy.
| |
Collapse
|
48
|
Huang W, Lv T, Li H, Du S, Yang C, Yuan S. [Correlation of apolipoprotein AI, apolipoprotein B and their ratio with the severity of cerebral white matter lesions]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2019; 38:992-996. [PMID: 30187870 DOI: 10.3969/j.issn.1673-4254.2018.08.15] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
OBJECTIVE To investigate the correlation of apolipoprotein AI (ApoAI), ApoB, ApoB/ApoAI and the severity of brain white matter lesions (WML). METHODS A total of 648 patients with WML confirmed by brain magnetic resonance imaging (MRI) were divided into mild WML group (n=386) and moderate to severe WML group (n=262) according to evaluations with the Fazekas scale. The demographic data, blood biochemical parameters and the levels of ApoAI, ApoB and ApoB/AI ratio were compared between the two groups to identify the risk factors of moderate to severe WML. RESULTS Univariate analysis showed that age, gender, hypertension, diabetes, coronary heart disease, previous stroke, homocysteine, HDL-C, ApoAI, and ApoB/AI ratio all differed significantly between the two groups (P < 0.05), but ApoB levels were similar between them (P > 0.05). Multivariate logistic regression analysis revealed that with ApoAI and ApoB/AI ratio as the continuous variables, after adjustment for the compounding factors, ApoB/AI ratio was an independent risk factor (OR=11.456, 95% CI: 3.622-36.229, P < 0.001) and ApoAI was an independent protective factor for moderate to severe WML (OR=0.068, 95% CI: 0.018-0.262, P < 0.001). With the upper quartiles of ApoAI level (1.38 g/L) and ApoB/AI ratio (0.58) as their respective cutoff values, patients with a high ApoAI level and a low ApoB/AI ratio were found to have the lowest incidence of moderate to severe WML (P < 0.001). CONCLUSIONS An increased ApoB/AI ratio is an independent risk factor and an increased ApoAI level is an independent protective factor for moderate to severe WML.
Collapse
Affiliation(s)
- Weihua Huang
- Department of Neurology, Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - Tianming Lv
- Department of Neurology, Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - Huanmin Li
- Department of Neurology, Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - Shuhua Du
- Department of Neurology, Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - Canhong Yang
- Department of Neurology, Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - Shiqi Yuan
- Department of Neurology, Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| |
Collapse
|
49
|
Mao L, Yang T, Li X, Lei X, Sun Y, Zhao Y, Zhang W, Gao Y, Sun B, Zhang F. Protective effects of sulforaphane in experimental vascular cognitive impairment: Contribution of the Nrf2 pathway. J Cereb Blood Flow Metab 2019; 39. [PMID: 29533123 PMCID: PMC6365596 DOI: 10.1177/0271678x18764083] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The major pathophysiological process of vascular cognitive impairment (VCI) is chronic cerebral ischemia, which causes disintegration of the blood-brain barrier (BBB), neuronal death, and white matter injury. This study aims to test whether sulforaphane (Sfn), a natural activator of nuclear factor erythroid 2-related factor 2 (Nrf2), reduces the chronic ischemic injury and cognitive dysfunction after VCI. Experimental VCI was induced in rats by permanent occlusion of both common carotid arteries for six weeks. This procedure caused notable neuronal death in the cortex and hippocampal CA1, myelin loss in the corpus callosum and hippocampal fimbria, accumulation of myelin debris in the corpus callosum, and remarkable cognitive impairment. Sfn treatment alleviated these ischemic injuries and the cognitive dysfunction. Sfn-mediated neuroprotection was associated with enhanced activation of Nrf2 and upregulation of heme oxygenase 1. Sfn also reduced neuronal and endothelial death and maintained the integrity of BBB after oxygen-glucose deprivation in vitro in an Nrf2 dependent manner. Furthermore, Nrf2 knockdown in endothelial cells decreased claudin-5 protein expression with downregulated claudin-5 promoter activity, suggesting that claudin-5 might be a target gene of Nrf2. Our results demonstrate that Sfn provides robust neuroprotection against chronic brain ischemic injury and may be a promising agent for VCI treatment.
Collapse
Affiliation(s)
- Leilei Mao
- 1 Department of Neurology, School of Medicine, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, Pittsburgh, PA, USA.,2 Key Laboratory of Cerebral Microcirculation, Taishan Medical University, Tai'an, Shandong, China
| | - Tuo Yang
- 1 Department of Neurology, School of Medicine, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xin Li
- 2 Key Laboratory of Cerebral Microcirculation, Taishan Medical University, Tai'an, Shandong, China
| | - Xia Lei
- 2 Key Laboratory of Cerebral Microcirculation, Taishan Medical University, Tai'an, Shandong, China
| | - Yang Sun
- 1 Department of Neurology, School of Medicine, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yongfang Zhao
- 3 State Key Laboratory of Medical Neurobiology and Institute of Brain Science, Fudan University, Shanghai, China
| | - Wenting Zhang
- 3 State Key Laboratory of Medical Neurobiology and Institute of Brain Science, Fudan University, Shanghai, China
| | - Yanqin Gao
- 1 Department of Neurology, School of Medicine, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, Pittsburgh, PA, USA.,3 State Key Laboratory of Medical Neurobiology and Institute of Brain Science, Fudan University, Shanghai, China
| | - Baoliang Sun
- 2 Key Laboratory of Cerebral Microcirculation, Taishan Medical University, Tai'an, Shandong, China
| | - Feng Zhang
- 1 Department of Neurology, School of Medicine, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, Pittsburgh, PA, USA.,2 Key Laboratory of Cerebral Microcirculation, Taishan Medical University, Tai'an, Shandong, China
| |
Collapse
|
50
|
Koundal S, Liu X, Sanggaard S, Mortensen K, Wardlaw J, Nedergaard M, Benveniste H, Lee H. Brain Morphometry and Longitudinal Relaxation Time of Spontaneously Hypertensive Rats (SHRs) in Early and Intermediate Stages of Hypertension Investigated by 3D VFA-SPGR MRI. Neuroscience 2019; 404:14-26. [PMID: 30690138 DOI: 10.1016/j.neuroscience.2019.01.030] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 01/15/2019] [Accepted: 01/18/2019] [Indexed: 02/03/2023]
Abstract
Cerebral small vessel disease(s) (SVD) results from pathological changes of the small blood vessels in the brain and is common in older people. The diagnostic features by which SVD manifests in brain includes white matter hyperintensities, lacunes, dilated perivascular spaces, microbleeds, and atrophy. In the present study, we use in vivo magnetic resonance imaging (MRI) to characterize brain morphometry and longitudinal relaxation time (T1) of spontaneously hypertensive rats (SHRs) to study the contribution of chronic hypertension to SVD relevant pathology. Male SHR and Wistar-Kyoto (WKY) rats underwent 3D variable flip angle spoiled gradient echo brain MRI at 9.4 T at early (seven weeks old) and established (19 weeks old) stages of hypertension. The derived proton density weighted and T1 images were utilized for morphometry and to characterize T1 properties in gray matter (GM), white matter (WM) and cerebrospinal fluid (CSF). Custom tissue probability maps were constructed for accurate computerized whole brain tissue segmentations and voxel-wise analyses. Characteristic morphological differences between the two strains included enlarged ventricles, smaller corpus callosum (CC) volumes and general 'thinning' of CC in SHR compared to WKY rats at both age groups. While we did not observe parenchymal T1 differences, the T1 of CSF was elevated in SHR compared to controls. Collectively these findings indicate that SHRs develop WM atrophy which is a clinically robust MRI biomarker associated with WM degeneration.
Collapse
Affiliation(s)
- Sunil Koundal
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, United States of America
| | - Xiaodan Liu
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, United States of America
| | - Simon Sanggaard
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, United States of America
| | - Kristian Mortensen
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Joanna Wardlaw
- Center for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK; UK Dementia Research Institute at The University of Edinburgh, The University of Edinburgh, Edinburgh, UK; Row Fogo Centre for Research into Ageing and the Brain, The University of Edinburgh, Edinburgh, UK
| | - Maiken Nedergaard
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Division of Glia Disease and Therapeutics, Center for Translational Neuromedicine, University of Rochester Medical School, Rochester, NY, USA
| | - Helene Benveniste
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, United States of America
| | - Hedok Lee
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, United States of America.
| |
Collapse
|