1
|
Pacella J, Lembo G, Carnevale L. A Translational Perspective on the Interplay Between Hypertension, Inflammation and Cognitive Impairment. Can J Cardiol 2024:S0828-282X(24)01033-X. [PMID: 39455022 DOI: 10.1016/j.cjca.2024.10.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 09/26/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024] Open
Abstract
Hypertension represents the major risk factor in the onset of cardiovascular disease worldwide. Preclinically, several mouse models of hypertension have been developed to investigate the pathophysiological link between hypertension and vascular impairment. Specifically, angiotensin-II infusion, transverse aortic constriction, deoxycorticosterone acetate salt, and L-NAME administration as hypertensive stimuli at the preclinical level permit the unveiling of a proinflammatory response driven by the innate and adaptive immune system and leads to vascular injury in terms of structural and functional alterations. Vascular impairment seems to be particularly critical at the cerebral level wherein arterioles, venules, and capillaries finely tune blood supply across the whole brain leading to the onset of a well known clinical condition named cerebral small vessel disease (cSVD) characterized by extensive brain injury, which culminates in the decline of cognitive functions. Advances in magnetic resonance imaging permit identification and accurate diagnosis of specific cSVD biomarkers including white matter hyperintensities, lacunar strokes, cerebral microbleeds, and enlarged perivascular spaces, each of which proved to be associated with a specific cognitive domain impairment. Such an approach in combination with pharmacological interventions targeted to the lowering of blood pressure and the prevention of vascular thrombosis formation represents a solid strategy in the prevention and the management of cSVD cognitive decay.
Collapse
Affiliation(s)
- Jacopo Pacella
- Department of Angiocardioneurology and Translational Medicine, IRCCS INM Neuromed, Pozzilli, Italy
| | - Giuseppe Lembo
- Department of Angiocardioneurology and Translational Medicine, IRCCS INM Neuromed, Pozzilli, Italy; Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy.
| | - Lorenzo Carnevale
- Department of Angiocardioneurology and Translational Medicine, IRCCS INM Neuromed, Pozzilli, Italy
| |
Collapse
|
2
|
Lin L, Chen Y, He K, Metwally S, Jha R, Capuk O, Bhuiyan MIH, Singh G, Cao G, Yin Y, Sun D. Carotid artery vascular stenosis causes the blood-CSF barrier damage and neuroinflammation. J Neuroinflammation 2024; 21:220. [PMID: 39256783 PMCID: PMC11385148 DOI: 10.1186/s12974-024-03209-1] [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: 03/20/2024] [Accepted: 08/26/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND The choroid plexus (ChP) helps maintain the homeostasis of the brain by forming the blood-CSF barrier via tight junctions (TJ) at the choroid plexus epithelial cells, and subsequently preventing neuroinflammation by restricting immune cells infiltration into the central nervous system. However, whether chronic cerebral hypoperfusion causes ChP structural damage and blood-CSF barrier impairment remains understudied. METHODS The bilateral carotid stenosis (BCAS) model in adult male C57BL/6 J mice was used to induce cerebral hypoperfusion, a model for vascular contributions to cognitive impairment and dementia (VCID). BCAS-mediated changes of the blood-CSF barrier TJ proteins, apical secretory Na+-K+-Cl- cotransporter isoform 1 (NKCC1) protein and regulatory serine-threonine kinases SPAK, and brain infiltration of myeloid-derived immune cells were assessed. RESULTS BCAS triggered dynamic changes of TJ proteins (claudin 1, claudin 5) accompanied with stimulation of SPAK-NKCC1 complex and NF-κB in the ChP epithelial cells. These changes impacted the integrity of the blood-CSF barrier, as evidenced by ChP infiltration of macrophages/microglia, neutrophils and T cells. Importantly, pharmacological blockade of SPAK with its potent inhibitor ZT1a in BCAS mice attenuated brain immune cell infiltration and improved cognitive neurological function. CONCLUSIONS BCAS causes chronic ChP blood-CSF damage and immune cell infiltration. Our study sheds light on the SPAK-NKCC1 complex as a therapeutic target in neuroinflammation.
Collapse
Affiliation(s)
- Lin Lin
- Department of Neurology, The Second Hospital of Dalian Medical University, Dalian, 116027, Liaoning, China
- Department of Neurology, University of Pittsburgh Medical Center, 7016 Biomedical Science Tower 3, 3501 Fifth Ave., Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yang Chen
- Department of Neurology, University of Pittsburgh Medical Center, 7016 Biomedical Science Tower 3, 3501 Fifth Ave., Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kai He
- Department of Neurology, University of Pittsburgh Medical Center, 7016 Biomedical Science Tower 3, 3501 Fifth Ave., Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA, USA
| | - Shamseldin Metwally
- Department of Neurology, University of Pittsburgh Medical Center, 7016 Biomedical Science Tower 3, 3501 Fifth Ave., Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA, USA
| | - Roshani Jha
- Department of Neurology, University of Pittsburgh Medical Center, 7016 Biomedical Science Tower 3, 3501 Fifth Ave., Pittsburgh, PA, 15213, USA
| | - Okan Capuk
- Department of Neurology, University of Pittsburgh Medical Center, 7016 Biomedical Science Tower 3, 3501 Fifth Ave., Pittsburgh, PA, 15213, USA
- Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Gazal Singh
- Biomedical Masters Program, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Guodong Cao
- Department of Neurology, University of Pittsburgh Medical Center, 7016 Biomedical Science Tower 3, 3501 Fifth Ave., Pittsburgh, PA, 15213, USA
- Research Service, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA
| | - Yan Yin
- Department of Neurology, The Second Hospital of Dalian Medical University, Dalian, 116027, Liaoning, China.
| | - Dandan Sun
- Department of Neurology, University of Pittsburgh Medical Center, 7016 Biomedical Science Tower 3, 3501 Fifth Ave., Pittsburgh, PA, 15213, USA.
- Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA, USA.
- Research Service, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA.
| |
Collapse
|
3
|
Biose IJ, Chastain WH, Solch-Ottaiano RJ, Grayson VS, Wang H, Banerjee S, Bix GJ. The Effects of Physical Activity on Experimental Models of Vascular Dementia: A Systematic Review and Meta-Analysis. Ann Neurosci 2024; 31:204-224. [PMID: 39156626 PMCID: PMC11325693 DOI: 10.1177/09727531231192759] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 06/16/2023] [Indexed: 08/20/2024] Open
Abstract
Background Physical activity is associated with improved brain health and cognition in humans. However, the validity, range, and quality of evidence for the beneficial outcomes linked to exercise in experimental models of vascular dementia (VaD) have not been evaluated. We performed a systematic review and meta-analysis of studies that assessed the effect of exercise intervention on models of VaD to provide an unbiased and comprehensive determination of the cognitive function and brain morphology benefits of exercise. Summary A systematic search in three databases as well as study design characteristics and experimental data extraction were completed in December 2021. We investigated the effects of exercise on cognitive function and brain-morphology outcomes in VaD models. Twenty-five studies were included for systematic review, while 21 studies were included in the meta-analysis. These studies included seven models of VaD in rats (60%, 15 studies), mice (36%, 9 studies), and pigs (4%, 1 study). None of the included studies used aged animals, and the majority of studies (80%) used only male animals. Key Message Exercise improves cognition but increased neuro-inflammation in VaD models Exercise improved cognitive function as well as some markers of brain morphology in models of VaD. However, exercise increased anxiety and neuro-inflammatory signals in VaD models. Further, we observed increased reporting anomalies such as a lack of blinding to group treatment or data analysis and randomization of animals to groups. Our report could help in the appropriate design of experimental studies seeking to investigate the effects of exercise as a non-pharmacological intervention on VaD models with a high translational impact.
Collapse
Affiliation(s)
- Ifechukwude J. Biose
- Department of Pharmacology and Experimental Therapeutics, Cardiovascular Center of Excellence, LSU Health Sciences Center, New Orleans, LA, USA
| | | | - Rebecca J. Solch-Ottaiano
- Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane University School of Medicine, New Orleans, LA, USA
| | | | - Hanyun Wang
- Department of Pharmacology and Experimental Therapeutics, Cardiovascular Center of Excellence, LSU Health Sciences Center, New Orleans, LA, USA
| | | | - Gregory J. Bix
- Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane University School of Medicine, New Orleans, LA, USA
- Tulane Brain Institute, Tulane University, New Orleans, LA, USA
- Department of Neurology, Tulane University School of Medicine, New Orleans, LA, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA
- Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| |
Collapse
|
4
|
Wu CYC, Zhang Y, Xu L, Huang Z, Zou P, Clemons GA, Li C, Citadin CT, Zhang Q, Lee RHC. The role of serum/glucocorticoid-regulated kinase 1 in brain function following cerebral ischemia. J Cereb Blood Flow Metab 2024; 44:1145-1162. [PMID: 38235747 PMCID: PMC11179613 DOI: 10.1177/0271678x231224508] [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: 08/15/2023] [Revised: 11/30/2023] [Accepted: 12/05/2023] [Indexed: 01/19/2024]
Abstract
Cardiopulmonary arrest (CA) is a major cause of death/disability in the U.S. with poor prognosis and survival rates. Current therapeutic challenges are physiologically complex because they involve hypoperfusion (decreased cerebral blood flow), neuroinflammation, and mitochondrial dysfunction. We previously discovered novel serum/glucocorticoid-regulated kinase 1 (SGK1) is highly expressed in brain of neurons that are susceptible to ischemia (hippocampus and cortex). We inhibited SGK1 and utilized pharmacological (specific inhibitor, GSK650394) and neuron-specific genetic approaches (shRNA) in rodent models of CA to determine if SGK1 is responsible for hypoperfusion, neuroinflammation, mitochondrial dysfunctional, and neurological deficits after CA. Inhibition of SGK1 alleviated cortical hypoperfusion and neuroinflammation (via Iba1, GFAP, and cytokine array). Treatment with GSK650394 enhanced mitochondrial function (via Seahorse respirometry) in the hippocampus 3 and 7 days after CA. Neuronal injury (via MAP2, dMBP, and Golgi staining) in the hippocampus and cortex was observed 7 days after CA but ameliorated with SGK1-shRNA. Moreover, SGK1 mediated neuronal injury by regulating the Ndrg1-SOX10 axis. Finally, animals subjected to CA exhibited learning/memory, motor, and anxiety deficits after CA, whereas SGK1 inhibition via SGK1-shRNA improved neurocognitive function. The present study suggests the fundamental roles of SGK1 in brain circulation and neuronal survival/death in cerebral ischemia-related diseases.
Collapse
Affiliation(s)
- Celeste Yin-Chieh Wu
- Stroke Center for Research, Louisiana State University Health, Shreveport, LA, USA
- Department of Neurology, Louisiana State University Health, Shreveport, LA, USA
| | - Yulan Zhang
- Stroke Center for Research, Louisiana State University Health, Shreveport, LA, USA
- Department of Neurology, Louisiana State University Health, Shreveport, LA, USA
| | - Li Xu
- Stroke Center for Research, Louisiana State University Health, Shreveport, LA, USA
- Department of Neurology, Louisiana State University Health, Shreveport, LA, USA
| | - Zhihai Huang
- Stroke Center for Research, Louisiana State University Health, Shreveport, LA, USA
- Department of Neurology, Louisiana State University Health, Shreveport, LA, USA
| | - Peibin Zou
- Stroke Center for Research, Louisiana State University Health, Shreveport, LA, USA
- Department of Neurology, Louisiana State University Health, Shreveport, LA, USA
| | - Garrett A Clemons
- Department of Cellular Biology and Anatomy, Louisiana State University Health, Shreveport, LA, USA
| | - Chun Li
- Stroke Center for Research, Louisiana State University Health, Shreveport, LA, USA
- Department of Neurology, Louisiana State University Health, Shreveport, LA, USA
| | - Cristiane T Citadin
- Department of Cellular Biology and Anatomy, Louisiana State University Health, Shreveport, LA, USA
| | - Quanguang Zhang
- Stroke Center for Research, Louisiana State University Health, Shreveport, LA, USA
- Department of Neurology, Louisiana State University Health, Shreveport, LA, USA
| | - Reggie Hui-Chao Lee
- Stroke Center for Research, Louisiana State University Health, Shreveport, LA, USA
- Department of Neurology, Louisiana State University Health, Shreveport, LA, USA
| |
Collapse
|
5
|
Wang L, Li M, Liu B, Zheng R, Zhang X, Yu S. miR-30a-5p mediates ferroptosis of hippocampal neurons in chronic cerebral hypoperfusion-induced cognitive dysfunction by modulating the SIRT1/NRF2 pathway. Brain Res Bull 2024; 212:110953. [PMID: 38636610 DOI: 10.1016/j.brainresbull.2024.110953] [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: 01/29/2024] [Revised: 04/07/2024] [Accepted: 04/16/2024] [Indexed: 04/20/2024]
Abstract
OBJECTIVE Chronic cerebral hypoperfusion (CCH) is a common cause of brain dysfunction. As a microRNA (also known as miRNAs or miRs), miR-30a-5p participates in neuronal damage and relates to ferroptosis. We explored the in vivo and in vitro effects and functional mechanism of miR-30a-5p in CCH-triggered cognitive impairment through the silent information regulator 1 (SIRT1)/nuclear factor erythroid 2-related factor 2 (NRF2) pathway. METHODS After 1 month of CCH modeling through bilateral common carotid artery stenosis, mice were injected with 2 μL antagomir (also known as anti-miRNAs) miR-30a-5p, with cognitive function evaluated by Morris water maze and novel object recognition tests. In vitro HT-22 cell oxygen glucose deprivation (OGD) model was established, followed by miR-30a-5p inhibitor and/or si-SIRT1 transfections, with Fe2+ concentration, malonaldehyde (MDA) and glutathione (GSH) contents, reactive oxygen species (ROS), miR-30a-5p and SIRT1 and glutathione peroxidase 4 (GPX4) protein levels, NRF2 nuclear translocation, and miR-30a-5p-SIRT1 targeting relationship assessed. RESULTS CCH-induced mice showed obvious cognitive impairment, up-regulated miR-30a-5p, and down-regulated SIRT1. Ferroptosis occurred in hippocampal neurons, manifested by elevated Fe2+ concentration and ROS and MDA levels, mitochondrial atrophy, and diminished GSH content. Antagomir miR-30a-5p or miR-30a-5p inhibitor promoted SIRT1 expression and NRF2 nuclear translocation, increased GPX4, cell viability and GSH content, and reduced Fe2+ concentration and ROS and MDA levels. miR-30a-5p negatively regulated SIRT1. In vitro, miR-30a-5p knockout increased NRF2 nuclear translocation by up-regulating SIRT1, inhibiting OGD-induced ferroptosis in HT-22 cells. CONCLUSION miR-30a-5p induces hippocampal neuronal ferroptosis and exacerbates post-CCH cognitive dysfunction by targeting SIRT1 and reducing NRF2 nuclear translocation.
Collapse
Affiliation(s)
- Lihua Wang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin, Heilongjiang 150086, China.
| | - Mingjie Li
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin, Heilongjiang 150086, China
| | - Bing Liu
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin, Heilongjiang 150086, China
| | - Ruihan Zheng
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin, Heilongjiang 150086, China
| | - Xinyi Zhang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin, Heilongjiang 150086, China
| | - Shuoyi Yu
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin, Heilongjiang 150086, China
| |
Collapse
|
6
|
Wu Y, Ke J, Ye S, Shan LL, Xu S, Guo SF, Li MT, Qiao TC, Peng ZY, Wang YL, Liu MY, Wang H, Feng JF, Han Y. 3D Visualization of Whole Brain Vessels and Quantification of Vascular Pathology in a Chronic Hypoperfusion Model Causing White Matter Damage. Transl Stroke Res 2024; 15:659-671. [PMID: 37222915 DOI: 10.1007/s12975-023-01157-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 05/02/2023] [Accepted: 05/11/2023] [Indexed: 05/25/2023]
Abstract
Chronic cerebral hypoperfusion is an important pathological factor in many neurodegenerative diseases, such as cerebral small vessel disease (CSVD). One of the most used animal models for chronic cerebral hypoperfusion is the bilateral common carotid artery stenosis (BCAS) mouse. For the therapy of CSVD and other diseases, it will be beneficial to understand the pathological alterations of the BCAS mouse, particularly vascular pathological changes. A mouse model of BCAS was used, and 8 weeks later, cognitive function of the mice was examined by using novel object recognition test and eight-arm radial maze test. 11.7 T magnetic resonance imaging (MRI) and luxol fast blue staining were used to evaluate the injury of the corpus callosum (CC), anterior commissure (AC), internal capsule (IC), and optic tract (Opt) in the cerebral white matter of mice. Three-dimensional vascular images of the whole brain of mice were acquired using fluorescence micro-optical sectioning tomography (fMOST) with a high resolution of 0.32 × 0.32 × 1.00 μm3. Then, the damaged white matter regions were further extracted to analyze the vessel length density, volume fraction, tortuosity, and the number of vessels of different internal diameters. The mouse cerebral caudal rhinal vein was also extracted and analyzed for its branch number and divergent angle in this study. BCAS modeling for 8 weeks resulted in impaired spatial working memory, reduced brain white matter integrity, and myelin degradation in mice, and CC showed the most severe white matter damage. 3D revascularization of the whole mouse brain showed that the number of large vessels was reduced and the number of small vessels was increased in BCAS mice. Further analysis revealed that the vessel length density and volume fraction in the damaged white matter region of BCAS mice were significantly reduced, and the vascular lesions were most noticeable in the CC. At the same time, the number of small vessels in the above white matter regions was significantly reduced, while the number of microvessels was significantly increased in BCAS mice, and the vascular tortuosity was also significantly increased. In addition, the analysis of caudal rhinal vein extraction revealed that the number of branches and the average divergent angle in BCAS mice were significantly reduced. The BCAS modeling for 8 weeks will lead to vascular lesions in whole brain of mice, and the caudal nasal vein was also damaged, while BCAS mice mainly mitigated the damages by increasing microvessels. What is more, the vascular lesions in white matter of mouse brain can cause white matter damage and spatial working memory deficit. These results provide evidence for the vascular pathological alterations caused by chronic hypoperfusion.
Collapse
Affiliation(s)
- Yang Wu
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai, 200437, China
| | - Jia Ke
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai, 200437, China
| | - Song Ye
- Wuhan OE-Bio Co., Ltd., G2 zone, Future City 999, Gaoxin boulevard East Lake High-Tech Development zone, Wuhan, 430074, China
| | - Li-Li Shan
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai, 200437, China
| | - Shuai Xu
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, 825 Zhangheng Road, Shanghai, 200127, China
| | - Shu-Fen Guo
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai, 200437, China
| | - Meng-Ting Li
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai, 200437, China
| | - Tian-Ci Qiao
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai, 200437, China
| | - Zheng-Yu Peng
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai, 200437, China
| | - Yi-Lin Wang
- Georgetown Preparatory School, Washington, DC, USA
| | - Ming-Yuan Liu
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai, 200437, China
| | - He Wang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, 825 Zhangheng Road, Shanghai, 200127, China.
| | - Jian-Feng Feng
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, 825 Zhangheng Road, Shanghai, 200127, China.
| | - Yan Han
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, 110 Ganhe Road, Shanghai, 200437, China.
| |
Collapse
|
7
|
Li D, Zhang Q, Yang X, Zhang G, Wang J, Zhang R, Liu Y. Microglial AT1R Conditional Knockout Ameliorates Hypoperfusive Cognitive Impairment by Reducing Microglial Inflammatory Responses. Neuroscience 2024; 545:125-140. [PMID: 38484837 DOI: 10.1016/j.neuroscience.2024.02.002] [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: 06/17/2023] [Revised: 02/01/2024] [Accepted: 02/05/2024] [Indexed: 03/24/2024]
Abstract
Chronic cerebral hypoperfusion (CCH) can cause vascular cognitive impairment and dementia. AT1R, angiotensin II type I receptor, plays a vital role in central nervous system pathologies, but its concrete function in vascular dementia is still unclear. Herein, we investigated the effects of AT1R during CCH by conditional knockout of the microglial AT1R and candesartan treatment. Using the bilateral carotid artery stenosis (BCAS) model, we found that the AT1R is crucial in exacerbating CCH-induced cognitive impairment via regulating microglial activation. The levels of AT1R were increased in the hippocampus and the hippocampal microglia after CCH induction. Microglial AT1R conditional knockout ameliorated cognitive impairment by reducing inflammatory responses and microglial activation, and so did candesartan treatment. However, we observed restoration of cerebral blood flow (CBF) but no significant neuronal loss in the hippocampus at 28 days after BCAS. Finally, we screened three hub genes (Ctss, Fcer1g, Tyrobp) associated with CCH. Our findings indicated that microglial expression of AT1R is critical for regulating neuroinflammation in CCH, and AT1R antagonism may be a feasible and promising method for ameliorating CCH-caused cognitive impairment.
Collapse
Affiliation(s)
- Deyue Li
- Department of Pharmacy, The Second Affiliated (Xinqiao) Hospital, The Army (Third Military) Medical University, Chongqing, China
| | - Qiao Zhang
- Department of Pain and Rehabilitation, The Second Affiliated (Xinqiao) Hospital, The Army (Third Military) Medical University, Chongqing, China
| | - Xia Yang
- Department of Wound Infection and Drug, State Key Laboratory of Trauma, Burn and Combined Injury, The Third Affiliated (Daping) Hospital, The Army (Third Military) Medical University, Chongqing, China
| | - Guoqing Zhang
- Department of Neurology, The Second Affiliated (Xinqiao) Hospital, The Army (Third Military) Medical University, Chongqing, China
| | - Jinping Wang
- Department of Neurology, The Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing, China
| | - Rong Zhang
- Department of Pharmacy, The Second Affiliated (Xinqiao) Hospital, The Army (Third Military) Medical University, Chongqing, China.
| | - Yong Liu
- Department of Pain and Rehabilitation, The Second Affiliated (Xinqiao) Hospital, The Army (Third Military) Medical University, Chongqing, China.
| |
Collapse
|
8
|
Zhang Z, Jin P, Guo Z, Tu Z, Yang H, Hu M, Li Q, Liu X, Li W, Hou S. Integrated Analysis of Chromatin and Transcriptomic Profiling Identifies PU.1 as a Core Regulatory Factor in Microglial Activation Induced by Chronic Cerebral Hypoperfusion. Mol Neurobiol 2024; 61:2569-2589. [PMID: 37917300 PMCID: PMC11043206 DOI: 10.1007/s12035-023-03734-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 10/20/2023] [Indexed: 11/04/2023]
Abstract
In addition to causing white matter lesions, chronic cerebral hypoperfusion (CCH) can also cause damage to gray matter, but the underlying molecular mechanisms remain largely unknown. In order to obtain a better understanding of the relationship between gene expression and transcriptional regulation alterations, novel upstream regulators could be identified using integration analysis of the transcriptome and epigenetic approaches. Here, a bilateral common carotid artery stenosis (BCAS) model was established for inducing CCH in mice. The spatial cognitive function of mice was evaluated, and changes in cortical microglia morphology were observed. RNA-sequencing (RNA-seq) and the assay for transposase-accessible chromatin with high-throughput sequencing (ATAC-seq) were performed on isolated mouse cortical brain tissue. Then, a systematic joint analysis of BCAS hypoperfusion-induced cortex-specific RNA-seq and ATAC-seq was conducted in order to assess the extent of the correlation between the two, and PU.1 was found to be greatly enriched through motif analysis and transcription factor annotation. Also, the core regulatory factor PU.1 induced by BCAS hypoperfusion was shown to be colocalized with microglia. Based on the above analysis, PU.1 plays a key regulatory role in microglial activation induced by CCH. And the transcriptome and epigenomic data presented in this study can help identify potential targets for future research exploring chronic hypoperfusion-induced brain injury.
Collapse
Affiliation(s)
- Zengyu Zhang
- Department of Neurology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Pengpeng Jin
- Department of Chronic Disease Management, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Zimin Guo
- Department of Neurology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhilan Tu
- Department of Neurology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Hualan Yang
- Department of Neurology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Mengting Hu
- Department of Neurology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Qinghua Li
- Department of Neurology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Xingdang Liu
- Department of Nuclear Medicine, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Weiwei Li
- Institute of Pediatrics, Children's Hospital of Fudan University, Fudan University, Shanghai, China.
| | - Shuangxing Hou
- Department of Neurology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China.
| |
Collapse
|
9
|
Wang Z, Han B, Qi J, Cao X, Gu H, Sun J. Chuanzhitongluo capsule improves cognitive impairment in mice with chronic cerebral hypoperfusion via the cholinergic anti-inflammatory pathway. Exp Gerontol 2024; 189:112407. [PMID: 38522309 DOI: 10.1016/j.exger.2024.112407] [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: 01/31/2024] [Revised: 03/08/2024] [Accepted: 03/19/2024] [Indexed: 03/26/2024]
Abstract
Vascular cognitive impairment (VCI) has become a common disease-causing cognitive deficit in humans, second only to Alzheimer's Disease (AD). Chuanzhitongluo capsule (CZTL) is a Traditional Chinese Medicine (TCM) preparation known for its effective protection against cerebral ischemia. However, its potential to ameliorate VCI remains unclear. This study aimed to investigate the cognitive improvement effects of CZTL in a mouse model of VCI. Chronic cerebral hypoperfusion (CCH) was induced in mice by bilateral common carotid artery stenosis (BCAS) to simulate the pathological changes associated with VCI. Spatial learning and memory abilities were assessed using the Morris Water Maze (MWM). RNA sequencing (RNA-Seq) was employed to identify differentially expressed genes (DEGs) in the hippocampus. Levels of inflammatory factors were measured through enzyme-linked immunosorbent assay (ELISA), while immunofluorescence (IF) determined the expression intensity of target proteins. Western Blot (WB) confirmed the final action pathway. Results indicated that CZTL significantly improved the spatial learning and memory abilities of CCH mice, along with alterations in gene expression profiles in the hippocampus. It also reduced neuroinflammation in the hippocampus and upregulated the choline acetyltransferase (ChAT) and α7 subunit-containing nicotinic acetylcholine receptor (α7nAChR), which are in synaptic plasticity and neuronal development. Moreover, CZTL inhibited the NF-κB signaling pathway. In conclusion, CZTL may alleviate neuroinflammation induced by CCH and improve cognitive impairment in CCH mice by regulating the cholinergic anti-inflammatory pathway (CAIP) involving ChAT/α7nAChR/NF-κB.
Collapse
Affiliation(s)
- Zhiyuan Wang
- Institute of Integrative Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Bin Han
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jianjiao Qi
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xuelei Cao
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Huali Gu
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Jinping Sun
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
10
|
Liu JA, Bumgarner JR, Walker WH, Meléndez-Fernández OH, Walton JC, DeVries AC, Nelson RJ. Chronic phase advances reduces recognition memory and increases vascular cognitive dementia-like impairments in aged mice. Sci Rep 2024; 14:7760. [PMID: 38565934 PMCID: PMC10987525 DOI: 10.1038/s41598-024-57511-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 03/19/2024] [Indexed: 04/04/2024] Open
Abstract
Disrupted or atypical light-dark cycles disrupts synchronization of endogenous circadian clocks to the external environment; extensive circadian rhythm desynchrony promotes adverse health outcomes. Previous studies suggest that disrupted circadian rhythms promote neuroinflammation and neuronal damage post-ischemia in otherwise healthy mice, however, few studies to date have evaluated these health risks with aging. Because most strokes occur in aged individuals, we sought to identify whether, in addition to being a risk factor for poor ischemic outcome, circadian rhythm disruption can increase risk for vascular cognitive impairment and dementia (VCID). We hypothesized that repeated 6 h phase advances (chronic jet lag; CJL) for 8 weeks alters cerebrovascular architecture leading to increased cognitive impairments in aged mice. Female CJL mice displayed impaired spatial processing during a spontaneous alternation task and reduced acquisition during auditory-cued associative learning. Male CJL mice displayed impaired retention of the auditory-cued associative learning task 24 h following acquisition. CJL increased vascular tortuosity in the isocortex, associated with increased risk for vascular disease. These results demonstrate that CJL increased sex-specific cognitive impairments coinciding with structural changes to vasculature in the brain. We highlight that CJL may accelerate aged-related functional decline and could be a crucial target against disease progression.
Collapse
Affiliation(s)
- Jennifer A Liu
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, USA.
| | - Jacob R Bumgarner
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, USA
| | - William H Walker
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, USA
- Department of Medicine, West Virginia University, Morgantown, USA
| | | | - James C Walton
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, USA
| | - A Courtney DeVries
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, USA
- Department of Medicine, West Virginia University, Morgantown, USA
- West Virginia University Cancer Institute, West Virginia University, Morgantown, USA
| | - Randy J Nelson
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, USA
| |
Collapse
|
11
|
Tian M, Kawaguchi R, Shen Y, Machnicki M, Villegas NG, Cooper DR, Montgomery N, Haring J, Lan R, Yuan AH, Williams CK, Magaki S, Vinters HV, Zhang Y, De Biase LM, Silva AJ, Carmichael ST. Intercellular Signaling Pathways as Therapeutic Targets for Vascular Dementia Repair. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.24.585301. [PMID: 38585718 PMCID: PMC10996514 DOI: 10.1101/2024.03.24.585301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Vascular dementia (VaD) is a white matter ischemic disease and the second-leading cause of dementia, with no direct therapy. Within the lesion site, cell-cell interactions dictate the trajectory towards disease progression or repair. To elucidate the underlying intercellular signaling pathways, a VaD mouse model was developed for transcriptomic and functional studies. The mouse VaD transcriptome was integrated with a human VaD snRNA-Seq dataset. A custom-made database encompassing 4053 human and 2032 mouse ligand-receptor (L-R) interactions identified significantly altered pathways shared between human and mouse VaD. Two intercellular L-R systems, Serpine2-Lrp1 and CD39-A3AR, were selected for mechanistic study as both the ligand and receptor were dysregulated in VaD. Decreased Seprine2 expression enhances OPC differentiation in VaD repair. A clinically relevant drug that reverses the loss of CD39-A3AR function promotes tissue and behavioral recovery in the VaD model. This study presents novel intercellular signaling targets and may open new avenues for VaD therapies.
Collapse
|
12
|
Xiao HH, Zhang FR, Li S, Guo FF, Hou JL, Wang SC, Yu J, Li XY, Yang HJ. Xinshubao tablet rescues cognitive dysfunction in a mouse model of vascular dementia: Involvement of neurogenesis and neuroinflammation. Biomed Pharmacother 2024; 172:116219. [PMID: 38310654 DOI: 10.1016/j.biopha.2024.116219] [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: 09/17/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/06/2024] Open
Abstract
Vascular dementia (VaD) represents a severe cognitive dysfunction syndrome closed linked to cardiovascular function. In the present study, we assessed the potential of Xinshubao tablet (XSB), a traditional Chinese prescription widely used for cardiovascular diseases, to mitigate neuropathological damage in a mouse model of VaD and elucidated the underlying mechanisms. Our findings revealed that oral administration of XSB rescued the cardiac dysfunction resulting from bilateral common carotid artery stenosis (BCAS), improved the cerebral blood flow (CBF) and cognitive function, reduced white matter injury, inhibited excessive microglial and astrocytic activation, stimulated hippocampal neurogenesis, and reduced neural apoptosis in the brains of BCAS mice. Mechanistically, RNA-seq analysis indicated that XSB treatment was significantly associated with neuroinflammation, vasculature development, and synaptic transmission, which were further confirmed by q-PCR assays. Western blot results revealed that XSB treatment hindered the nuclear translocation of nuclear factor-κB (NF-κB), thereby suppressing the NF-κB signaling pathway. These results collectively demonstrated that XSB could ameliorate cognitive dysfunction caused by BCAS through regulating CBF, reducing white matter lesions, suppressing glial activation, promoting neurogenesis, and mitigating neuroinflammation. Notably, the NF-κB signaling pathway emerged as a pivotal player in this mechanism.
Collapse
Affiliation(s)
- Hong-He Xiao
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China; School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China; Fujian Pien Tze Huang Enterprise Key Laboratory of Natural Medicine Research and Development, Zhangzhou Pien Tze Huang Pharmaceutical Co., Ltd., Zhangzhou, Fujian Province 363099, China
| | - Feng-Rong Zhang
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China; Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Sen Li
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Fei-Fei Guo
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Jin-Li Hou
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Shi-Cong Wang
- Fujian Pien Tze Huang Enterprise Key Laboratory of Natural Medicine Research and Development, Zhangzhou Pien Tze Huang Pharmaceutical Co., Ltd., Zhangzhou, Fujian Province 363099, China
| | - Juan Yu
- Fujian Pien Tze Huang Enterprise Key Laboratory of Natural Medicine Research and Development, Zhangzhou Pien Tze Huang Pharmaceutical Co., Ltd., Zhangzhou, Fujian Province 363099, China.
| | - Xian-Yu Li
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Hong-Jun Yang
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China; China Academy of Chinese Medical Sciences, Beijing 100700, China.
| |
Collapse
|
13
|
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
|
14
|
Dinh QN, Arumugam T. The Bilateral Carotid Artery Stenosis (BCAS) Model of Vascular Dementia. Methods Mol Biol 2024; 2746:67-72. [PMID: 38070080 DOI: 10.1007/978-1-0716-3585-8_5] [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: 12/18/2023]
Abstract
Vascular dementia is the second most common form of dementia after Alzheimer's disease. Chronic cerebral hypoperfusion is a key contributor to the development of vascular dementia. In this chapter, we describe the surgical procedures used for bilateral carotid artery stenosis (BCAS) surgery to induce chronic cerebral hypoperfusion. Mice that undergo BCAS surgery develop the hallmarks of vascular dementia including white matter lesions, neuroinflammation, and cognitive impairment. This technique may be used for studies of chronic cerebral hypoperfusion and vascular dementia in mice.
Collapse
Affiliation(s)
- Quynh Nhu Dinh
- Department of Microbiology, Anatomy, Physiology and Pharmacology, Centre for Cardiovascular Biology and Disease Research, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia.
| | - Thiruma Arumugam
- Department of Microbiology, Anatomy, Physiology and Pharmacology, Centre for Cardiovascular Biology and Disease Research, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia
| |
Collapse
|
15
|
Saito S, Suzuki K, Ohtani R, Maki T, Kowa H, Tachibana H, Washida K, Kawabata N, Mizuno T, Kanki R, Sudoh S, Kitaguchi H, Shindo K, Shindo A, Oka N, Yamamoto K, Yasuno F, Kakuta C, Kakuta R, Yamamoto Y, Hattori Y, Takahashi Y, Nakaoku Y, Tonomura S, Oishi N, Aso T, Taguchi A, Kagimura T, Kojima S, Taketsuna M, Tomimoto H, Takahashi R, Fukuyama H, Nagatsuka K, Yamamoto H, Fukushima M, Ihara M. Efficacy and Safety of Cilostazol in Mild Cognitive Impairment: A Randomized Clinical Trial. JAMA Netw Open 2023; 6:e2344938. [PMID: 38048134 PMCID: PMC10696485 DOI: 10.1001/jamanetworkopen.2023.44938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 10/15/2023] [Indexed: 12/05/2023] Open
Abstract
Importance Recent evidence indicates the efficacy of β-amyloid immunotherapy for the treatment of Alzheimer disease, highlighting the need to promote β-amyloid removal from the brain. Cilostazol, a selective type 3 phosphodiesterase inhibitor, promotes such clearance by facilitating intramural periarterial drainage. Objective To determine the safety and efficacy of cilostazol in mild cognitive impairment. Design, Setting, and Participants The COMCID trial (A Trial of Cilostazol for Prevention of Conversion from Mild Cognitive Impairment to Dementia) was an investigator-initiated, double-blind, phase 2 randomized clinical trial. Adult participants were registered between May 25, 2015, and March 31, 2018, and received placebo or cilostazol for up to 96 weeks. Participants were treated in the National Cerebral and Cardiovascular Center and 14 other regional core hospitals in Japan. Patients with mild cognitive impairment with Mini-Mental State Examination (MMSE) scores of 22 to 28 points (on a scale of 0 to 30, with lower scores indicating greater cognitive impairment) and Clinical Dementia Rating scores of 0.5 points (on a scale of 0, 0.5, 1, 2, and 3, with higher scores indicating more severe dementia) were enrolled. The data were analyzed from May 1, 2020, to December 1, 2020. Interventions The participants were treated with placebo, 1 tablet twice daily, or cilostazol, 50 mg twice daily, for up to 96 weeks. Main Outcomes and Measures The primary end point was the change in the total MMSE score from baseline to the final observation. Safety analyses included all adverse events. Results The full analysis set included 159 patients (66 [41.5%] male; mean [SD] age, 75.6 [5.2] years) who received placebo or cilostazol at least once. There was no statistically significant difference between the placebo and cilostazol groups for the primary outcome. The least-squares mean (SE) changes in the MMSE scores among patients receiving placebo were -0.1 (0.3) at the 24-week visit, -0.8 (0.3) at 48 weeks, -1.2 (0.4) at 72 weeks, and -1.3 (0.4) at 96 weeks. Among those receiving cilostazol, the least-squares mean (SE) changes in MMSE scores were -0.6 (0.3) at 24 weeks, -1.0 (0.3) at 48 weeks, -1.1 (0.4) at 72 weeks, and -1.8 (0.4) at 96 weeks. Two patients (2.5%) in the placebo group and 3 patients (3.8%) in the cilostazol group withdrew owing to adverse effects. There was 1 case of subdural hematoma in the cilostazol group, which may have been related to the cilostazol treatment; the patient was successfully treated surgically. Conclusions and Relevance In this randomized clinical trial, cilostazol was well tolerated, although it did not prevent cognitive decline. The efficacy of cilostazol should be tested in future trials. Trial Registration ClinicalTrials.gov Identifier: NCT02491268.
Collapse
Affiliation(s)
- Satoshi Saito
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Keisuke Suzuki
- Innovation Center for Translational Research, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Ryo Ohtani
- Department of Neurology, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Takakuni Maki
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hisatomo Kowa
- Division of Neurology, Kobe University Hospital, Kobe, Japan
| | | | - Kazuo Washida
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Japan
| | | | - Toshiki Mizuno
- Department of Neurology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Rie Kanki
- Department of Neurology, Osaka City General Hospital, Osaka, Japan
| | - Shinji Sudoh
- Department of Neurology, National Hospital Organization, Utano National Hospital, Kyoto, Japan
| | - Hiroshi Kitaguchi
- Department of Neurology, Kurashiki Central Hospital, Kurashiki, Japan
| | - Katsuro Shindo
- Department of Neurology, Kurashiki Central Hospital, Kurashiki, Japan
| | - Akihiro Shindo
- Department of Neurology, Graduate School of Medicine, Mie University, Tsu, Japan
| | - Nobuyuki Oka
- Department of Neurology, National Hospital Organization Minami Kyoto Hospital, Joyo, Japan
| | - Keiichi Yamamoto
- Internal Medicine and Neurology, Nara Midori Clinic, Nara, Japan
| | - Fumihiko Yasuno
- Department of Psychiatry, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Chikage Kakuta
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Ryosuke Kakuta
- Department of Data Science, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Yumi Yamamoto
- Department of Molecular Innovation in Lipidemiology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Yorito Hattori
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Yukako Takahashi
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Yuriko Nakaoku
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shuichi Tonomura
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Naoya Oishi
- Department of Psychiatry, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Toshihiko Aso
- Laboratory for Brain Connectomics Imaging, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Akihiko Taguchi
- Department of Regenerative Medicine Research, Institute of Biomedical Research and Innovation, Kobe, Japan
| | - Tatsuo Kagimura
- Translational Research Center for Medical Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
| | - Shinsuke Kojima
- Translational Research Center for Medical Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
| | - Masanori Taketsuna
- Translational Research Center for Medical Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
| | - Hidekazu Tomimoto
- Department of Neurology, Graduate School of Medicine, Mie University, Tsu, Japan
| | - Ryosuke Takahashi
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hidenao Fukuyama
- Research and Educational Unit of Leaders for Integrated Medical System, Kyoto University, Kyoto, Japan
| | - Kazuyuki Nagatsuka
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Haruko Yamamoto
- Department of Data Science, National Cerebral and Cardiovascular Center, Suita, Japan
| | | | - Masafumi Ihara
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Japan
| |
Collapse
|
16
|
He Y, Li Z, Shi X, Ding J, Wang X. Metformin attenuates white matter injury and cognitive impairment induced by chronic cerebral hypoperfusion. J Cereb Blood Flow Metab 2023; 43:78-94. [PMID: 37177813 PMCID: PMC10638997 DOI: 10.1177/0271678x231175189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 03/28/2023] [Accepted: 04/01/2023] [Indexed: 05/15/2023]
Abstract
Vascular cognitive impairment and dementia (VCID) is a series of cognitive dysfunction associated with cerebrovascular diseases and currently lacks effective treatments. The white matter, which is essential for neuronal information processing and integration, is nourished by a network of capillaries and is vulnerable to chronic hypoperfusion. Here, we show that metformin, a widely used drug for the treatment of type 2 diabetes, alleviates the white matter damage and improves cognitive impairment in a mouse model of VCID established by bilateral carotid artery stenosis (BCAS)-induced chronic hypoperfusion. Mechanistically, metformin restores the dysfunctions of oligodendrocyte precursor cells (OPCs) under hypoxia. Metformin up-regulates prolyl hydroxylases 2 via activating the AMP-activated protein kinase pathway, leading to hypoxia-inducible factor-1α (HIF-1α) degradation in OPCs. These findings suggest that metformin may have a promising therapeutic role in alleviating cognitive abnormalities by ameliorating white matter damage of VCID.
Collapse
Affiliation(s)
- Yixi He
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Zhenghao Li
- Institute of Neuroscience, MOE Key Laboratory of Molecular Neurobiology, NMU, Shanghai, China
| | - Xiaoyu Shi
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Jing Ding
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Xin Wang
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
17
|
Ishikawa H, Shindo A, Mizutani A, Tomimoto H, Lo EH, Arai K. A brief overview of a mouse model of cerebral hypoperfusion by bilateral carotid artery stenosis. J Cereb Blood Flow Metab 2023; 43:18-36. [PMID: 36883344 PMCID: PMC10638994 DOI: 10.1177/0271678x231154597] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 12/23/2022] [Accepted: 01/04/2023] [Indexed: 03/09/2023]
Abstract
Vascular cognitive impairment (VCI) refers to all forms of cognitive disorder related to cerebrovascular diseases, including vascular mild cognitive impairment, post-stroke dementia, multi-infarct dementia, subcortical ischemic vascular dementia (SIVD), and mixed dementia. Among the causes of VCI, more attention has been paid to SIVD because the causative cerebral small vessel pathologies are frequently observed in elderly people and because the gradual progression of cognitive decline often mimics Alzheimer's disease. In most cases, small vessel diseases are accompanied by cerebral hypoperfusion. In mice, prolonged cerebral hypoperfusion is induced by bilateral carotid artery stenosis (BCAS) with surgically implanted metal micro-coils. This cerebral hypoperfusion BCAS model was proposed as a SIVD mouse model in 2004, and the spreading use of this mouse SIVD model has provided novel data regarding cognitive dysfunction and histological/genetic changes by cerebral hypoperfusion. Oxidative stress, microvascular injury, excitotoxicity, blood-brain barrier dysfunction, and secondary inflammation may be the main mechanisms of brain damage due to prolonged cerebral hypoperfusion, and some potential therapeutic targets for SIVD have been proposed by using transgenic mice or clinically used drugs in BCAS studies. This review article overviews findings from the studies that used this hypoperfused-SIVD mouse model, which were published between 2004 and 2021.
Collapse
Affiliation(s)
- Hidehiro Ishikawa
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Akihiro Shindo
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Akane Mizutani
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Hidekazu Tomimoto
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Eng H Lo
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Ken Arai
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| |
Collapse
|
18
|
Wang X, Chen W, Yuan P, Xu H. RAGE acted as a new anti-inflammatory target for Icariin's treatment against vascular dementia based on network pharmacology-directed verification. J Biomol Struct Dyn 2023; 42:10189-10209. [PMID: 37768122 DOI: 10.1080/07391102.2023.2256409] [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: 06/26/2023] [Accepted: 08/22/2023] [Indexed: 09/29/2023]
Abstract
Vascular dementia (VaD) ranks as the second most prevalent form of dementia and poses a considerable global health challenge. Icariin has been recognized for its robust neuroprotective effects in combating VaD. Nonetheless, the underlying mechanisms have not been fully elucidated. An integrated approach involving network pharmacology, molecular docking, and molecular dynamics simulations (MDS) was employed to systematically investigate the potential pharmacological actions of Icariin in counteracting VaD. The AGE/RAGE pathway was identified as a promising anti-inflammatory pathway. A chronic cerebral hypoperfusion mouse model was utilized to establish VaD. Both Icariin and FP S-ZM1 (a RAGE inhibitor) were administered through oral gavage and intraperitoneal injection, respectively. The Morris water maze (MWZ) was used to evaluate cognitive functions. Moreover, immunofluorescence, RT-qP CR, and Western blot analyses were carried out to evaluate the effects of FP S-ZM1 on neuroinflammation. Network analysis identified 14 crucial targets and highlighted the AGE-RAGE signaling cascade in diabetic complications as the foremost KEGG pathway with potential anti-neuroinflammatory property. MDS results suggested a stable binding of the RAGE-Icariin complex. Remarkably, Icariin was found to effectively mitigate cognitive deficits in VaD mice, which was correlated with the upregulation of the P I3K/AKT pathway and downregulation of the JNK/cJUN signaling cascade. Critically, co-administration of FP S-ZM1 enhanced Icariin's ameliorative effects on cognitive deficits, owing to bolstered anti-neuroinflammatory action. This study unveils the potential of Icariin in alleviating cognitive dysfunction and neuroinflammation in VaD, which may be attributed to the modulation of the AGE/RAGE pathway.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Xiaohu Wang
- Department of Neurology, The Affiliated Hospital of Guizhou Medical University, Guizhou, China
| | - Wei Chen
- Department of Neurology, The Affiliated Hospital of Guizhou Medical University, Guizhou, China
| | - Ping Yuan
- Department of Neurology, The Affiliated Hospital of Guizhou Medical University, Guizhou, China
| | - Hongbei Xu
- Department of Neurology, The Affiliated Hospital of Guizhou Medical University, Guizhou, China
| |
Collapse
|
19
|
Fraga E, Medina V, Cuartero MI, García-Culebras A, Bravo-Ferrer I, Hernández-Jiménez M, Garcia-Segura JM, Hurtado O, Pradillo JM, Lizasoain I, Moro MÁ. Defective hippocampal neurogenesis underlies cognitive impairment by carotid stenosis-induced cerebral hypoperfusion in mice. Front Cell Neurosci 2023; 17:1219847. [PMID: 37636586 PMCID: PMC10457159 DOI: 10.3389/fncel.2023.1219847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 07/31/2023] [Indexed: 08/29/2023] Open
Abstract
Chronic cerebral hypoperfusion due to carotid artery stenosis is a major cause of vascular cognitive impairment and dementia (VCID). Bilateral carotid artery stenosis (BCAS) in rodents is a well-established model of VCID where most studies have focused on white matter pathology and subsequent cognitive deficit. Therefore, our aim was to study the implication of adult hippocampal neurogenesis in hypoperfusion-induced VCID in mice, and its relationship with cognitive hippocampal deficits. Mice were subjected to BCAS; 1 and 3 months later, hippocampal memory and neurogenesis/cell death were assessed, respectively, by the novel object location (NOL) and spontaneous alternation performance (SAP) tests and by immunohistology. Hypoperfusion was assessed by arterial spin labeling-magnetic resonance imaging (ASL-MRI). Hypoperfused mice displayed spatial memory deficits with decreased NOL recognition index. Along with the cognitive deficit, a reduced number of newborn neurons and their aberrant morphology indicated a remarkable impairment of the hippocampal neurogenesis. Both increased cell death in the subgranular zone (SGZ) and reduced neuroblast proliferation rate may account for newborn neurons number reduction. Our data demonstrate quantitative and qualitative impairment of adult hippocampal neurogenesis disturbances associated with cerebral hypoperfusion-cognitive deficits in mice. These findings pave the way for novel diagnostic and therapeutic targets for VCID.
Collapse
Affiliation(s)
- Enrique Fraga
- Neurovascular Pathophysiology Group, Cardiovascular Risk Factor and Brain Function Programme, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Unidad de Investigación Neurovascular, Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
- Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - Violeta Medina
- Neurovascular Pathophysiology Group, Cardiovascular Risk Factor and Brain Function Programme, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Unidad de Investigación Neurovascular, Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
- Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - María Isabel Cuartero
- Neurovascular Pathophysiology Group, Cardiovascular Risk Factor and Brain Function Programme, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Unidad de Investigación Neurovascular, Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
- Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - Alicia García-Culebras
- Neurovascular Pathophysiology Group, Cardiovascular Risk Factor and Brain Function Programme, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Unidad de Investigación Neurovascular, Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
- Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid (UCM), Madrid, Spain
- Departamento de Biología Celular, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - Isabel Bravo-Ferrer
- Unidad de Investigación Neurovascular, Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - Macarena Hernández-Jiménez
- Unidad de Investigación Neurovascular, Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - Juan Manuel Garcia-Segura
- Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid (UCM), Madrid, Spain
- ICTS Bioimagen Complutense, Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - Olivia Hurtado
- Unidad de Investigación Neurovascular, Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - Jesus Miguel Pradillo
- Unidad de Investigación Neurovascular, Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
- Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - Ignacio Lizasoain
- Unidad de Investigación Neurovascular, Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
- Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - María Ángeles Moro
- Neurovascular Pathophysiology Group, Cardiovascular Risk Factor and Brain Function Programme, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Unidad de Investigación Neurovascular, Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid (UCM), Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| |
Collapse
|
20
|
Manukjan N, Majcher D, Leenders P, Caiment F, van Herwijnen M, Smeets HJ, Suidgeest E, van der Weerd L, Vanmierlo T, Jansen JFA, Backes WH, van Oostenbrugge RJ, Staals J, Fulton D, Ahmed Z, Blankesteijn WM, Foulquier S. Hypoxic oligodendrocyte precursor cell-derived VEGFA is associated with blood-brain barrier impairment. Acta Neuropathol Commun 2023; 11:128. [PMID: 37550790 PMCID: PMC10405482 DOI: 10.1186/s40478-023-01627-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 07/23/2023] [Indexed: 08/09/2023] Open
Abstract
Cerebral small vessel disease is characterised by decreased cerebral blood flow and blood-brain barrier impairments which play a key role in the development of white matter lesions. We hypothesised that cerebral hypoperfusion causes local hypoxia, affecting oligodendrocyte precursor cell-endothelial cell signalling leading to blood-brain barrier dysfunction as an early mechanism for the development of white matter lesions. Bilateral carotid artery stenosis was used as a mouse model for cerebral hypoperfusion. Pimonidazole, a hypoxic cell marker, was injected prior to humane sacrifice at day 7. Myelin content, vascular density, blood-brain barrier leakages, and hypoxic cell density were quantified. Primary mouse oligodendrocyte precursor cells were exposed to hypoxia and RNA sequencing was performed. Vegfa gene expression and protein secretion was examined in an oligodendrocyte precursor cell line exposed to hypoxia. Additionally, human blood plasma VEGFA levels were measured and correlated to blood-brain barrier permeability in normal-appearing white matter and white matter lesions of cerebral small vessel disease patients and controls. Cerebral blood flow was reduced in the stenosis mice, with an increase in hypoxic cell number and blood-brain barrier leakages in the cortical areas but no changes in myelin content or vascular density. Vegfa upregulation was identified in hypoxic oligodendrocyte precursor cells, which was mediated via Hif1α and Epas1. In humans, VEGFA plasma levels were increased in patients versus controls. VEGFA plasma levels were associated with increased blood-brain barrier permeability in normal appearing white matter of patients. Cerebral hypoperfusion mediates hypoxia induced VEGFA expression in oligodendrocyte precursor cells through Hif1α/Epas1 signalling. VEGFA could in turn increase BBB permeability. In humans, increased VEGFA plasma levels in cerebral small vessel disease patients were associated with increased blood-brain barrier permeability in the normal appearing white matter. Our results support a role of VEGFA expression in cerebral hypoperfusion as seen in cerebral small vessel disease.
Collapse
Affiliation(s)
- Narek Manukjan
- Department of Pharmacology and Toxicology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- CARIM - School for Cardiovascular Diseases, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Edgbaston, Birmingham, B15 2TT UK
| | - Daria Majcher
- Department of Pharmacology and Toxicology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Peter Leenders
- Department of Pharmacology and Toxicology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Florian Caiment
- Department of Toxicogenomics, GROW–School for Oncology and Developmental Biology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Marcel van Herwijnen
- Department of Toxicogenomics, GROW–School for Oncology and Developmental Biology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Hubert J. Smeets
- Department of Toxicogenomics, GROW–School for Oncology and Developmental Biology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- MHeNs—School for Mental Health and Neuroscience, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Ernst Suidgeest
- C.J. Gorter Center for High Field MRI, Department of Radiology, Leiden University Medical Center, P.O. Box 9500, 2300 RA Leiden, the Netherlands
| | - Louise van der Weerd
- C.J. Gorter Center for High Field MRI, Department of Radiology, Leiden University Medical Center, P.O. Box 9500, 2300 RA Leiden, the Netherlands
- Department of Human Genetics, Leiden University Medical Center, P.O. Box 9500, 2300 RA Leiden, The Netherlands
| | - Tim Vanmierlo
- MHeNs—School for Mental Health and Neuroscience, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- Department of Neuroscience, Biomedical Research Institute, Hasselt University, 3500 Hasselt, Belgium
- Department of Psychiatry and Neuropsychology, European Graduate School of Neuroscience, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Jacobus F. A. Jansen
- MHeNs—School for Mental Health and Neuroscience, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | - Walter H. Backes
- CARIM - School for Cardiovascular Diseases, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- MHeNs—School for Mental Health and Neuroscience, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | - Robert J. van Oostenbrugge
- CARIM - School for Cardiovascular Diseases, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- MHeNs—School for Mental Health and Neuroscience, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- Department of Neurology, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | - Julie Staals
- CARIM - School for Cardiovascular Diseases, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- Department of Neurology, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | - Daniel Fulton
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Edgbaston, Birmingham, B15 2TT UK
| | - Zubair Ahmed
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Edgbaston, Birmingham, B15 2TT UK
- Centre for Trauma Sciences Research, University of Birmingham, Edgbaston, Birmingham, B15 2TT UK
| | - W. Matthijs Blankesteijn
- Department of Pharmacology and Toxicology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- CARIM - School for Cardiovascular Diseases, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Sébastien Foulquier
- Department of Pharmacology and Toxicology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- CARIM - School for Cardiovascular Diseases, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- MHeNs—School for Mental Health and Neuroscience, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- Department of Neurology, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| |
Collapse
|
21
|
Perrotta M, Carnevale D, Carnevale L. Mouse models of cerebral injury and cognitive impairment in hypertension. Front Aging Neurosci 2023; 15:1199612. [PMID: 37539342 PMCID: PMC10394515 DOI: 10.3389/fnagi.2023.1199612] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 07/05/2023] [Indexed: 08/05/2023] Open
Abstract
Hypertension is a major risk factor for dementia, including both vascular and neurodegenerative etiologies. With the original aim of studying the effect of blood pressure elevation on canonical target organs of hypertension as the heart, the vasculature or the kidneys, several experimental models of hypertension have sprouted during the years. With the more recent interest of understanding the cerebral injury burden caused by hypertension, it is worth understanding how the main models of hypertension or localized cerebral hypertension stand in the field of hypertension-induced cerebral injury and cognitive impairment. With this review we will report main genetic, pharmacological and surgical models of cognitive impairment induced by hypertension, summarizing how each specific category and model can improve our understanding of the complex phenomenon of cognitive loss of vascular etiology.
Collapse
Affiliation(s)
- Marialuisa Perrotta
- Department of Molecular Medicine, “Sapienza” University of Rome, Rome, Italy
- Research Unit of Neuro and Cardiovascular Pathophysiology, IRCCS Neuromed, Department of Angiocardioneurology and Translational Medicine, Pozzilli, Italy
| | - Daniela Carnevale
- Department of Molecular Medicine, “Sapienza” University of Rome, Rome, Italy
- Research Unit of Neuro and Cardiovascular Pathophysiology, IRCCS Neuromed, Department of Angiocardioneurology and Translational Medicine, Pozzilli, Italy
| | - Lorenzo Carnevale
- Research Unit of Neuro and Cardiovascular Pathophysiology, IRCCS Neuromed, Department of Angiocardioneurology and Translational Medicine, Pozzilli, Italy
| |
Collapse
|
22
|
Zhang Y, Tan J, Yang K, Fan W, Yu B, Shi W. Ambient RNAs removal of cortex-specific snRNA-seq reveals Apoe + microglia/macrophage after deeper cerebral hypoperfusion in mice. J Neuroinflammation 2023; 20:152. [PMID: 37365617 DOI: 10.1186/s12974-023-02831-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 06/12/2023] [Indexed: 06/28/2023] Open
Abstract
BACKGROUND Ambient RNAs contamination in single-nuclei RNA sequencing (snRNA-seq) is a challenging problem, but the consequences of ambient RNAs contamination of damaged and/or diseased tissues are poorly understood. Cognitive impairments and white/gray matter injuries are characteristic of deeper cerebral hypoperfusion mouse models induced by bilateral carotid artery stenosis (BCAS), but the molecular mechanisms still need to be further explored. More importantly, the BCAS mice can also offer an excellent model to examine the signatures of ambient RNAs contamination in damaged tissues when performing snRNA-seq. METHODS After the sham and BCAS mice were established, cortex-specific single-nuclei libraries were constructed. Single-nuclei transcriptomes were described informatically by the R package Seurat, and ambient RNA markers of were identified in each library. Then, after removing ambient RNAs in each sample using the in silico approaches, the combination of CellBender and subcluster cleaning, single-nuclei transcriptomes were reconstructed. Next, the comparison of ambient RNA contamination was performed using irGSEA analysis before and after the in silico approaches. Finally, further bioinformatic analyses were performed. RESULTS The ambient RNAs are more predominant in the BCAS group than the sham group. The contamination mainly originated from damaged neuronal nuclei, but could be reduced largely using the in silico approaches. The integrative analysis of cortex-specific snRNA-seq data and the published bulk transcriptome revealed that microglia and other immune cells were the primary effectors. In the sequential microglia/immune subgroups analysis, the subgroup of Apoe+ MG/Mac (microglia/macrophages) was identified. Interestingly, this subgroup mainly participated in the pathways of lipid metabolism, associated with the phagocytosis of cell debris. CONCLUSIONS Taken together, our current study unravels the features of ambient RNAs in snRNA-seq datasets under diseased conditions, and the in silico approaches can effectively eliminate the incorrected cell annotation and following misleading analysis. In the future, snRNA-seq data analysis should be carefully revisited, and ambient RNAs removal needs to be taken into consideration, especially for those diseased tissues. To our best knowledge, our study also offers the first cortex-specific snRNA-seq data of deeper cerebral hypoperfusion, which provides with novel therapeutic targets.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Vascular Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, People's Republic of China
- Fudan Zhangjiang Institute, Shanghai, 201203, China
| | - Jinyun Tan
- Department of Vascular Surgery, Huashan Hospital of Fudan University, Shanghai, People's Republic of China
| | - Kai Yang
- Department of Vascular Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, People's Republic of China
| | - Weijian Fan
- Department of Vascular Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, People's Republic of China
| | - Bo Yu
- Department of Vascular Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, People's Republic of China.
- Fudan Zhangjiang Institute, Shanghai, 201203, China.
| | - Weihao Shi
- Department of Vascular Surgery, Huashan Hospital of Fudan University, Shanghai, People's Republic of China.
| |
Collapse
|
23
|
Rajeev V, Chai YL, Poh L, Selvaraji S, Fann DY, Jo DG, De Silva TM, Drummond GR, Sobey CG, Arumugam TV, Chen CP, Lai MKP. Chronic cerebral hypoperfusion: a critical feature in unravelling the etiology of vascular cognitive impairment. Acta Neuropathol Commun 2023; 11:93. [PMID: 37309012 PMCID: PMC10259064 DOI: 10.1186/s40478-023-01590-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 05/25/2023] [Indexed: 06/14/2023] Open
Abstract
Vascular cognitive impairment (VCI) describes a wide spectrum of cognitive deficits related to cerebrovascular diseases. Although the loss of blood flow to cortical regions critically involved in cognitive processes must feature as the main driver of VCI, the underlying mechanisms and interactions with related disease processes remain to be fully elucidated. Recent clinical studies of cerebral blood flow measurements have supported the role of chronic cerebral hypoperfusion (CCH) as a major driver of the vascular pathology and clinical manifestations of VCI. Here we review the pathophysiological mechanisms as well as neuropathological changes of CCH. Potential interventional strategies for VCI are also reviewed. A deeper understanding of how CCH can lead to accumulation of VCI-associated pathology could potentially pave the way for early detection and development of disease-modifying therapies, thus allowing preventive interventions instead of symptomatic treatments.
Collapse
Affiliation(s)
- Vismitha Rajeev
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore
| | - Yuek Ling Chai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore
| | - Luting Poh
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore
| | - Sharmelee Selvaraji
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore
- Integrative Sciences and Engineering Programme, NUS Graduate School, National University of Singapore, Singapore, Singapore
| | - David Y Fann
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - T Michael De Silva
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia
| | - Grant R Drummond
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia
| | - Christopher G Sobey
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia
| | - Thiruma V Arumugam
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia
| | - Christopher P Chen
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore
- NUS Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Mitchell K P Lai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore.
- NUS Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
24
|
Zhang Z, Guo Z, Jin P, Yang H, Hu M, Zhang Y, Tu Z, Hou S. Transcriptome Profiling of Hippocampus After Cerebral Hypoperfusion in Mice. J Mol Neurosci 2023; 73:423-436. [PMID: 37266840 PMCID: PMC10432347 DOI: 10.1007/s12031-023-02123-0] [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: 02/10/2023] [Accepted: 05/18/2023] [Indexed: 06/03/2023]
Abstract
Chronic cerebral hypoperfusion (CCH) is considered to be one of the major mechanism in the pathogenesis of vascular cognitive impairment (VCI). Increased inflammatory cells, particularly microglia, often parallel hypoperfusion-induced gray matter damage such as hippocampal lesions, but the exact mechanism remains largely unknown. To understand the pathological mechanisms, we analyzed hippocampus-specific transcriptome profiles after cerebral hypoperfusion. The mouse hypoperfusion model was induced by employing the 0.16/0.18 mm bilateral common carotid artery stenosis (BCAS) procedure. Cerebral blood flow (CBF) was assessed after 3-week hypoperfusion. Pathological changes were evaluated via hematoxylin staining and immunofluorescence staining. RNA-sequencing (RNA-seq) was performed using RNA samples of sham- or BCAS-operated mice, followed by quantitative real-time PCR (qRT-PCR) validation. We found that the 0.16/0.18 mm BCAS induced decreased CBF, hippocampal neuronal loss, and microglial activation. Furthermore, GSEA between sham and BCAS mice showed activation of interferon-beta signaling along with inflammatory immune responses. In addition, integrative analysis with published single-cell RNA-seq revealed that up-regulated differentially expressed genes (DEGs) were enriched in a distinct cell type of "microglia," and down-regulated DEGs were enriched in "CA1 pyramidal," not in "interneurons" or "S1 pyramidal." This database of transcriptomic profiles of BCAS-hypoperfusion will be useful for future studies to explore potential targets for vascular cognitive dysfunction.
Collapse
Affiliation(s)
- Zengyu Zhang
- Department of Neurology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
- Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Zimin Guo
- Department of Neurology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
- Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Pengpeng Jin
- Department of Chronic Disease Management, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Hualan Yang
- Department of Neurology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Mengting Hu
- Department of Neurology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Yuan Zhang
- Department of Vascular Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 201399, Shanghai, China
| | - Zhilan Tu
- Department of Neurology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China.
| | - Shuangxing Hou
- Department of Neurology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China.
| |
Collapse
|
25
|
Sampath D, Branyan TE, Markowsky KG, Gunda R, Samiya N, Obenaus A, Sohrabji F. Sex differences in cognitive impairment after focal ischemia in middle-aged rats and the effect of iv miR-20a-3p treatment. Neurobiol Aging 2023; 129:168-177. [PMID: 37336171 DOI: 10.1016/j.neurobiolaging.2023.05.001] [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: 01/22/2023] [Revised: 05/02/2023] [Accepted: 05/02/2023] [Indexed: 06/21/2023]
Abstract
Stroke is a major cause of death and disability worldwide and is also a leading cause of vascular dementia and Alzheimer's disease, with older women experiencing accelerated decline. Our previous studies show that intravenous (iv) injections of miR-20a-3p, a small noncoding RNA (miRNA) delivered after stroke improves acute stroke outcomes in middle-aged male and female rats. The present study tested whether mir-20a-3p treatment would also ameliorate stroke-induced cognitive decline in the chronic phase. Acyclic middle-aged females and age-matched male Sprague Dawley rats were subjected to middle cerebral artery occlusion using endothelin-1 or sham surgery, and treated iv with miR-20a-3p mimics or scrambled oligos at 4 hours, 24 hours, and 70 days post-stroke. Stroke resulted in a significant sensory motor deficit, while miR-20a-3p treatment reduced these deficits in both sexes. Cognitive impairment was assessed periodically for 3 months after stroke using contextual fear conditioning and the novel object recognition task. Overall, the tests of associative and episodic memory were affected by focal ischemia only in female rats, and miR-20a-3p ameliorated the rate of decline.
Collapse
Affiliation(s)
- Dayalan Sampath
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, College of Medicine and Texas A&M Institute for Neuroscience, Texas A&M University, Bryan, TX, USA
| | - Taylor E Branyan
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, College of Medicine and Texas A&M Institute for Neuroscience, Texas A&M University, Bryan, TX, USA
| | - Kylee G Markowsky
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, College of Medicine and Texas A&M Institute for Neuroscience, Texas A&M University, Bryan, TX, USA
| | - Rithvik Gunda
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, College of Medicine and Texas A&M Institute for Neuroscience, Texas A&M University, Bryan, TX, USA
| | - Nadia Samiya
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, College of Medicine and Texas A&M Institute for Neuroscience, Texas A&M University, Bryan, TX, USA
| | - Andre Obenaus
- Department of Pediatrics, University of California Irvine, Irvine, CA, USA
| | - Farida Sohrabji
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, College of Medicine and Texas A&M Institute for Neuroscience, Texas A&M University, Bryan, TX, USA.
| |
Collapse
|
26
|
Zhang Z, Guo Z, Tu Z, Yang H, Li C, Hu M, Zhang Y, Jin P, Hou S. Cortex-specific transcriptome profiling reveals upregulation of interferon-regulated genes after deeper cerebral hypoperfusion in mice. Front Physiol 2023; 14:1056354. [PMID: 36994418 PMCID: PMC10040763 DOI: 10.3389/fphys.2023.1056354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 02/24/2023] [Indexed: 03/16/2023] Open
Abstract
Background: Chronic cerebral hypoperfusion (CCH) is commonly accompanied by brain injury and glial activation. In addition to white matter lesions, the intensity of CCH greatly affects the degree of gray matter damage. However, little is understood about the underlying molecular mechanisms related to cortical lesions and glial activation following hypoperfusion. Efforts to investigate the relationship between neuropathological alternations and gene expression changes support a role for identifying novel molecular pathways by transcriptomic mechanisms.Methods: Chronic cerebral ischemic injury model was induced by the bilateral carotid artery stenosis (BCAS) using 0.16/0.18 mm microcoils. Cerebral blood flow (CBF) was evaluated using laser speckle contrast imaging (LSCI) system. Spatial learning and memory were assessed by Morris water maze test. Histological changes were evaluated by Hematoxylin staining. Microglial activation and neuronal loss were further examined by immunofluorescence staining. Cortex-specific gene expression profiling analysis was performed in sham and BCAS mice, and then validated by quantitative RT-PCR and immunohistochemistry (IHC).Results: In our study, compared with the sham group, the right hemisphere CBF of BCAS mice decreased to 69% and the cognitive function became impaired at 4 weeks postoperation. Besides, the BCAS mice displayed profound gray matter damage, including atrophy and thinning of the cortex, accompanied by neuronal loss and increased activated microglia. Gene set enrichment analysis (GSEA) revealed that hypoperfusion-induced upregulated genes were significantly enriched in the pathways of interferon (IFN)-regulated signaling along with neuroinflammation signaling. Ingenuity pathway analysis (IPA) predicted the importance of type I IFN signaling in regulating the CCH gene network. The obtained RNA-seq data were validated by qRT-PCR in cerebral cortex, showing consistency with the RNA-seq results. Also, IHC staining revealed elevated expression of IFN-inducible protein in cerebral cortex following BCAS-hypoperfusion.Conclusion: Overall, the activation of IFN-mediated signaling enhanced our understanding of the neuroimmune responses induced by CCH. The upregulation of IFN-regulated genes (IRGs) might exert a critical impact on the progression of cerebral hypoperfusion. Our improved understanding of cortex-specific transcriptional profiles will be helpful to explore potential targets for CCH.
Collapse
Affiliation(s)
- Zengyu Zhang
- Department of Neurology, Shanghai Pudong Hospital, Fudan University, Shanghai, China
| | - Zimin Guo
- Department of Neurology, Shanghai Pudong Hospital, Fudan University, Shanghai, China
| | - Zhilan Tu
- Department of Neurology, Shanghai Pudong Hospital, Fudan University, Shanghai, China
| | - Hualan Yang
- Department of Neurology, Shanghai Pudong Hospital, Fudan University, Shanghai, China
| | - Chao Li
- School of Pharmacy, Hubei University of Science and Technology, Hubei, China
| | - Mengting Hu
- Department of Neurology, Shanghai Pudong Hospital, Fudan University, Shanghai, China
| | - Yuan Zhang
- Department of Vascular Surgery, Shanghai Pudong Hospital, Fudan University, Shanghai, China
| | - Pengpeng Jin
- Department of Chronic Disease Management, Shanghai Pudong Hospital, Fudan University, Shanghai, China
- *Correspondence: Shuangxing Hou, ; Pengpeng Jin,
| | - Shuangxing Hou
- Department of Neurology, Shanghai Pudong Hospital, Fudan University, Shanghai, China
- *Correspondence: Shuangxing Hou, ; Pengpeng Jin,
| |
Collapse
|
27
|
Li J, Zou X, Mao R, Han L, Xia S, Yang H, Cao X, Xu Y. Determination of the role of hippocampal astrocytes in the bilateral common carotid artery stenosis mouse model by RNA sequencing. Neurosci Lett 2023; 805:137213. [PMID: 36966961 DOI: 10.1016/j.neulet.2023.137213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/21/2023] [Accepted: 03/22/2023] [Indexed: 04/07/2023]
Abstract
INTRODUCTION Bilateral common carotid artery stenosis (BCAS) is used experimentally to model vascular dementia (VaD). Previous studies have primarily focused on the degradation of brain white matter after BCAS. However, hippocampal abnormalities are equally important, and hippocampal astrocytes are specifically involved in neural circuits that regulate learning and memory. Whether hippocampal astrocytes participate in the pathogenesis of BCAS-induced VaD has not been well studied. Therefore, in the present study, we attempted to explore the role of hippocampal astrocytes in BCAS. METHODS Two months after BCAS, behavioral experiments were conducted to investigate changes in neurological function in sham and BCAS mice. A ribosome-tagging approach (RiboTag) profiling strategy was used to isolate mRNAs enriched in hippocampal astrocytes, and the RNA was sequenced and analyzed using transcriptomic methods. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) was utilized to validate the results of RNA sequencing. Immunofluorescence analyses were conducted to evaluate the number and morphology of hippocampal astrocytes. RESULTS We observed significant short-term working memory impairment in BCAS mice. Moreover, the RNA obtained through RiboTag technology was specific to astrocytes. Transcriptomics approaches and subsequent validation studies revealed that the genes that showed expression changes in hippocampal astrocytes after BCAS were mainly involved in immune system processes, glial cell proliferation, substance transport and metabolism. Furthermore, the number and distribution of astrocytes in the CA1 region of the hippocampus tended to decrease after modeling. CONCLUSION In this study, comparisons between sham and BCAS mice showed that the functions of hippocampal astrocytes were impaired in BCAS-induced chronic cerebral hypoperfusion-related VaD.
Collapse
Affiliation(s)
- Jiangnan Li
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu 210008, China
| | - Xinxin Zou
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Nanjing, Jiangsu 210008, China
| | - Rui Mao
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, Jiangsu 210008, China
| | - Lijian Han
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu 210008, China
| | - Shengnan Xia
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, Jiangsu 210008, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu 210008, China; Jiangsu Provincial Key Discipline of Neurology, Nanjing, Jiangsu 210008, China; Nanjing Neurology Medical Center, Nanjing, Jiangsu 210008, China
| | - Haiyan Yang
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, Jiangsu 210008, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu 210008, China; Jiangsu Provincial Key Discipline of Neurology, Nanjing, Jiangsu 210008, China; Nanjing Neurology Medical Center, Nanjing, Jiangsu 210008, China
| | - Xiang Cao
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu 210008, China; Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Nanjing, Jiangsu 210008, China; Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, Jiangsu 210008, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu 210008, China; Jiangsu Provincial Key Discipline of Neurology, Nanjing, Jiangsu 210008, China; Nanjing Neurology Medical Center, Nanjing, Jiangsu 210008, China.
| | - Yun Xu
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu 210008, China; Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Nanjing, Jiangsu 210008, China; Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, Jiangsu 210008, China; Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu 210008, China; Jiangsu Provincial Key Discipline of Neurology, Nanjing, Jiangsu 210008, China; Nanjing Neurology Medical Center, Nanjing, Jiangsu 210008, China.
| |
Collapse
|
28
|
Maki K, Ohara T, Hata J, Shibata M, Hirabayashi N, Honda T, Sakata S, Furuta Y, Akiyama M, Yamasaki K, Tatewaki Y, Taki Y, Kitazono T, Mikami T, Maeda T, Ono K, Mimura M, Nakashima K, Iga JI, Takebayashi M, Ninomiya T. CKD, Brain Atrophy, and White Matter Lesion Volume: The Japan Prospective Studies Collaboration for Aging and Dementia. Kidney Med 2023; 5:100593. [PMID: 36874508 PMCID: PMC9982615 DOI: 10.1016/j.xkme.2022.100593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 10/26/2022] [Indexed: 12/24/2022] Open
Abstract
Rationale & Objective Chronic kidney disease, defined by albuminuria and/or reduced estimated glomerular filtration rate (eGFR), has been reported to be associated with brain atrophy and/or higher white matter lesion volume (WMLV), but there are few large-scale population-based studies assessing this issue. This study aimed to examine the associations between the urinary albumin-creatinine ratio (UACR) and eGFR levels and brain atrophy and WMLV in a large-scale community-dwelling older population of Japanese. Study Design Population-based cross-sectional study. Setting & Participants A total of 8,630 dementia-free community-dwelling Japanese aged greater than or equal to 65 years underwent brain magnetic resonance imaging scanning and screening examination of health status in 2016-2018. Exposures UACR and eGFR levels. Outcomes The total brain volume (TBV)-to-intracranial volume (ICV) ratio (TBV/ICV), the regional brain volume-to-TBV ratio, and the WMLV-to-ICV ratio (WMLV/ICV). Analytical Approach The associations of UACR and eGFR levels with the TBV/ICV, the regional brain volume-to-TBV ratio, and the WMLV/ICV were assessed by using an analysis of covariance. Results Higher UACR levels were significantly associated with lower TBV/ICV and higher geometric mean values of the WMLV/ICV (P for trend = 0.009 and <0.001, respectively). Lower eGFR levels were significantly associated with lower TBV/ICV, but not clearly associated with WMLV/ICV. In addition, higher UACR levels, but not lower eGFR, were significantly associated with lower temporal cortex volume-to-TBV ratio and lower hippocampal volume-to-TBV ratio. Limitations Cross-sectional study, misclassification of UACR or eGFR levels, generalizability to other ethnicities and younger populations, and residual confounding factors. Conclusions The present study demonstrated that higher UACR was associated with brain atrophy, especially in the temporal cortex and hippocampus, and with increased WMLV. These findings suggest that chronic kidney disease is involved in the progression of morphologic brain changes associated with cognitive impairment.
Collapse
Affiliation(s)
- Kenji Maki
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomoyuki Ohara
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Jun Hata
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Center for Cohort Studies, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Mao Shibata
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Center for Cohort Studies, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Psychosomatic Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Naoki Hirabayashi
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Psychosomatic Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takanori Honda
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Satoko Sakata
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Center for Cohort Studies, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshihiko Furuta
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masato Akiyama
- Department of Ocular Pathology and Imaging Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Keisuke Yamasaki
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yasuko Tatewaki
- Department of Aging Research and Geriatric Medicine, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Yasuyuki Taki
- Department of Aging Research and Geriatric Medicine, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Takanari Kitazono
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Center for Cohort Studies, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tatsuya Mikami
- Department of Preemptive Medicine, Graduate School of Medicine, Hirosaki University, Hirosaki, Japan
| | - Tetsuya Maeda
- Division of Neurology and Gerontology, Department of Internal Medicine, School of Medicine, Iwate Medical University, Iwate, Japan
| | - Kenjiro Ono
- Department of Neurology, Kanazawa University Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Masaru Mimura
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Kenji Nakashima
- National Hospital Organization, Matsue Medical Center, Shimane, Japan
| | - Jun-Ichi Iga
- Department of Neuropsychiatry, Ehime University Graduate School of Medicine, Ehime University, Ehime, Japan
| | - Minoru Takebayashi
- Faculty of Life Sciences, Department of Neuropsychiatry, Kumamoto University, Kumamoto, Japan
| | - Toshiharu Ninomiya
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Center for Cohort Studies, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | |
Collapse
|
29
|
Che P, Zhang J, Yu M, Tang P, Wang Y, Lin A, Xu J, Zhang N. Dl-3-n-butylphthalide promotes synaptic plasticity by activating the Akt/ERK signaling pathway and reduces the blood-brain barrier leakage by inhibiting the HIF-1α/MMP signaling pathway in vascular dementia model mice. CNS Neurosci Ther 2023; 29:1392-1404. [PMID: 36756709 PMCID: PMC10068471 DOI: 10.1111/cns.14112] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 12/29/2022] [Accepted: 01/20/2023] [Indexed: 02/10/2023] Open
Abstract
AIMS DL-3-n-butylphthalide (NBP) exerts beneficial effects on global cognitive functions, but the underlying molecular mechanisms are still poorly understood. The present study aimed to investigate whether NBP mediates synaptic plasticity and blood-brain barrier (BBB) function, which play a pivotal role in the pathogenesis of vascular dementia (VaD), in a mouse model of bilateral common carotid artery stenosis (BCAS). METHODS NBP was administered to model mice at a dose of 80 mg/kg by gavage for 28 days after surgery. Cognitive function was evaluated by behavioral tests, and hippocampal synaptic plasticity was evaluated by in vivo electrophysiological recording. Cerebral blood flow (CBF), hippocampal volume, and white matter integrity were measured with laser speckle imaging (LSI) and MRI. In addition, BBB leakage and the expression of proteins related to the Akt/ERK and HIF-1α/MMP signaling pathways were assessed by biochemical assays. RESULTS NBP treatment alleviated cognitive impairment, hippocampal atrophy, and synaptic plasticity impairment induced by BCAS. In addition, NBP treatment increased CBF, promoted white matter integrity, and decreased BBB leakage. Regarding the molecular mechanisms, in mice with BCAS, NBP may activate the Akt/ERK signaling pathway, which upregulates the expression of synapse-associated proteins, and it may also inhibit the HIF-1α/MMP signaling pathway, thereby increasing the expression of tight junction (TJ) proteins. CONCLUSION In conclusion, our results demonstrated the therapeutic effects of NBP in improving cognitive function via a wide range of targets in mice subjected to BCAS.
Collapse
Affiliation(s)
- Ping Che
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Juan Zhang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China.,Department of Neurology, Gucheng Hospital in Hebei Province, Hengshui, China
| | - Mingqian Yu
- School of Medicine, Nankai University, Tianjin, China
| | - Ping Tang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Yanhui Wang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Aolei Lin
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Jing Xu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Nan Zhang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China.,Department of Neurology, Tianjin Medical University General Hospital Airport Site, Tianjin, China
| |
Collapse
|
30
|
Lin H, Zhang J, Dai Y, Liu H, He X, Chen L, Tao J, Li C, Liu W. Neurogranin as an important regulator in swimming training to improve the spatial memory dysfunction of mice with chronic cerebral hypoperfusion. JOURNAL OF SPORT AND HEALTH SCIENCE 2023; 12:116-129. [PMID: 35066217 PMCID: PMC9923430 DOI: 10.1016/j.jshs.2022.01.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 11/18/2021] [Accepted: 12/30/2021] [Indexed: 05/09/2023]
Abstract
BACKGROUND Vascular cognitive impairment caused by chronic cerebral hypoperfusion (CCH) has become a hot issue worldwide. Aerobic exercise positively contributes to the preservation or restoration of cognitive abilities; however, the specific mechanism has remained inconclusive. And recent studies found that neurogranin (Ng) is a potential biomarker for cognitive impairment. This study aims to investigate the underlying role of Ng in swimming training to improve cognitive impairment. METHODS To test this hypothesis, the clustered regularly interspaced short palindromic repeats (CRISPR)-associated protein 9 (Cas9) system was utilized to construct a strain of Ng conditional knockout (Ng cKO) mice, and bilateral common carotid artery stenosis (BCAS) surgery was performed to prepare the model. In Experiment 1, 2-month-old male and female transgenic mice were divided into a control group (wild-type littermate, n = 9) and a Ng cKO group (n = 9). Then, 2-month-old male and female C57BL/6 mice were divided into a sham group (C57BL/6, n = 12) and a BCAS group (n = 12). In Experiment 2, 2-month-old male and female mice were divided into a sham group (wild-type littermate, n = 12), BCAS group (n = 12), swim group (n = 12), BCAS + Ng cKO group (n = 12), and swim + Ng cKO group (n = 12). Then, 7 days after BCAS, mice were given swimming training for 5 weeks (1 week for adaptation and 4 weeks for training, 5 days a week, 60 min a day). After intervention, laser speckle was used to detect cerebral blood perfusion in the mice, and the T maze and Morris water maze were adopted to test their spatial memory. Furthermore, electrophysiology and Western blotting were conducted to record long-term potential and observe the expressions of Ca2+ pathway-related proteins, respectively. Immunohistochemistry was applied to analyze the expression of relevant markers in neuronal damage, inflammation, and white matter injury. RESULTS The figures showed that spatial memory impairment was detected in Ng cKO mice, and a sharp decline of cerebral blood flow and an impairment of progressive spatial memory were observed in BCAS mice. Regular swimming training improved the spatial memory impairment of BCAS mice. This was achieved by preventing long-term potential damage and reversing the decline of Ca2+ signal transduction pathway-related proteins. At the same time, the results suggested that swimming also led to improvements in neuronal death, inflammation, and white matter injury induced by CCH. Further study adopted the use of Ng cKO transgenic mice, and the results indicated that the positive effects of swimming training on cognitive impairments, synaptic plasticity, and related pathological changes caused by CCH could be abolished by the knockout of Ng. CONCLUSION Swimming training can mediate the expression of Ng to enhance hippocampal synaptic plasticity and improve related pathological changes induced by CCH, thereby ameliorating the spatial memory impairment of vascular cognitive impairment.
Collapse
Affiliation(s)
- Huawei Lin
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Jiayong Zhang
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Yaling Dai
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Huanhuan Liu
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Xiaojun He
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Lewen Chen
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Jing Tao
- Rehabilitation Industry Institute, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Chaohui Li
- General surgery, Anxi General Hospital of Traditional Chinese Medicine, Quanzhou 362400, China
| | - Weilin Liu
- Rehabilitation Industry Institute, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China.
| |
Collapse
|
31
|
He Y, Chen X, Wu M, Hou X, Zhou Z. What type of cell death occurs in chronic cerebral hypoperfusion? A review focusing on pyroptosis and its potential therapeutic implications. Front Cell Neurosci 2023; 17:1073511. [PMID: 36937182 PMCID: PMC10017988 DOI: 10.3389/fncel.2023.1073511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 01/31/2023] [Indexed: 03/06/2023] Open
Abstract
Chronic cerebral hypoperfusion (CCH) is a major global disease with chronic cerebral blood flow reduction. It is also the main cause of cognitive impairment and neurodegenerative diseases. Pyroptosis, a novel form of cell death, is characterized by the rupture of the cell membrane and the release of pro-inflammatory mediators. In recent years, an increasing number of studies have identified the involvement of pyroptosis and its mediated inflammatory response in the pathological process of CCH. Therefore, preventing the activation of pyroptosis following CCH is beneficial to inhibit the inflammatory cascade and reduce brain injury. In this review, we discuss the research progress on the relationship between pyroptosis and CCH, in order to provide a reference for research in related fields.
Collapse
Affiliation(s)
- Yuxuan He
- Department of Neurology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- Department of Neurology, School of Medicine, Chongqing University, Chongqing, China
| | - Xi Chen
- Department of Neurology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Min Wu
- Department of Neurology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xianhua Hou
- Department of Neurology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- *Correspondence: Xianhua Hou Zhenhua Zhou
| | - Zhenhua Zhou
- Department of Neurology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- *Correspondence: Xianhua Hou Zhenhua Zhou
| |
Collapse
|
32
|
Cognitive Impairments and blood-brain Barrier Damage in a Mouse Model of Chronic Cerebral Hypoperfusion. Neurochem Res 2022; 47:3817-3828. [DOI: 10.1007/s11064-022-03799-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 10/07/2022] [Accepted: 10/16/2022] [Indexed: 10/31/2022]
Abstract
AbstractChronic cerebral hypoperfusion (CCH) is commonly involved in various brain diseases. Tight junction proteins (TJs) are key components constituting the anatomical substrate of the blood-brain barrier (BBB). Changes in cognitive function and BBB after CCH and their relationship need further exploration. To investigate the effect of CCH on cognition and BBB, we developed a bilateral common carotid artery stenosis (BCAS) model in Tie2-GFP mice. Mice manifested cognitive impairments accompanied with increased microglia after the BCAS operation. BCAS mice also exhibited increased BBB permeability at all time points set from D1 to D42. Furthermore, BCAS mice showed reduced expression of TJs 42 d after the operation. In addition, correct entrances of mice in radial arm maze test had a moderate negative correlation with EB extravasation. Our data suggested that BCAS could lead to cognitive deficits, microglia increase and BBB dysfunction characterized by increased BBB permeability and reduced TJs expression level. BBB permeability may be involved in the cognitive impairments induced by CCH.
Collapse
|
33
|
Belmonte KCD, Holmgren EB, Wills TA, Gidday JM. Epigenetic conditioning induces intergenerational resilience to dementia in a mouse model of vascular cognitive impairment. Alzheimers Dement 2022; 18:1711-1720. [PMID: 35170835 PMCID: PMC9790554 DOI: 10.1002/alz.12616] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 12/10/2021] [Accepted: 01/10/2022] [Indexed: 01/28/2023]
Abstract
INTRODUCTION Epigenetic stimuli induce beneficial or detrimental changes in gene expression, and consequently, phenotype. Some of these phenotypes can manifest across the lifespan-and even in subsequent generations. Here, we used a mouse model of vascular cognitive impairment and dementia (VCID) to determine whether epigenetically induced resilience to specific dementia-related phenotypes is heritable by first-generation progeny. METHODS Our systemic epigenetic therapy consisted of 2 months of repetitive hypoxic "conditioning" (RHC) prior to chronic cerebral hypoperfusion in adult C57BL/6J mice. Resultant changes in object recognition memory and hippocampal long-term potentiation (LTP) were assessed 3 and 4 months later, respectively. RESULTS Hypoperfusion-induced memory/plasticity deficits were abrogated by RHC. Moreover, similarly robust dementia resilience was documented in untreated cerebral hypoperfused animals derived from RHC-treated parents. CONCLUSIONS Our results in experimental VCID underscore the efficacy of epigenetics-based treatments to prevent memory loss, and demonstrate for the first time the heritability of an induced resilience to dementia.
Collapse
Affiliation(s)
- Krystal Courtney D. Belmonte
- Department of OphthalmologyLouisiana State University School of MedicineLSUHSCNew OrleansLouisianaUSA,Department of PhysiologyLouisiana State University School of MedicineLSUHSCNew OrleansLouisianaUSA
| | - Eleanor B. Holmgren
- Department of Cell Biology and AnatomyLouisiana State University School of MedicineLSUHSCNew OrleansLouisianaUSA
| | - Tiffany A. Wills
- Department of Cell Biology and AnatomyLouisiana State University School of MedicineLSUHSCNew OrleansLouisianaUSA,Neuroscience Center of ExcellenceLouisiana State University School of MedicineLSUHSCNew OrleansLouisianaUSA
| | - Jeff M. Gidday
- Department of OphthalmologyLouisiana State University School of MedicineLSUHSCNew OrleansLouisianaUSA,Department of PhysiologyLouisiana State University School of MedicineLSUHSCNew OrleansLouisianaUSA,Neuroscience Center of ExcellenceLouisiana State University School of MedicineLSUHSCNew OrleansLouisianaUSA,Department of Biochemistry and Molecular BiologyLouisiana State University School of MedicineLSUHSCNew OrleansLouisianaUSA
| |
Collapse
|
34
|
Bhatia K, Kindelin A, Nadeem M, Khan MB, Yin J, Fuentes A, Miller K, Turner GH, Preul MC, Ahmad AS, Mufson EJ, Waters MF, Ahmad S, Ducruet AF. Complement C3a Receptor (C3aR) Mediates Vascular Dysfunction, Hippocampal Pathology, and Cognitive Impairment in a Mouse Model of VCID. Transl Stroke Res 2022; 13:816-829. [PMID: 35258803 DOI: 10.1007/s12975-022-00993-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 01/24/2022] [Accepted: 01/28/2022] [Indexed: 01/12/2023]
Abstract
Vascular contributions to cognitive impairment and dementia (VCID) secondary to chronic mild-moderate cerebral ischemia underlie a significant percentage of cases of dementia. We previously reported that either genetic deficiency of the complement C3a receptor (C3aR) or its pharmacological inhibition protects against cerebral ischemia in rodents, while others have implicated C3aR in the pathogenesis seen in rodent transgenic models of Alzheimer's disease. In the present study, we evaluated the role of complement C3a-C3aR signaling in the onset and progression of VCID. We utilized the bilateral common carotid artery stenosis (BCAS) model to induce VCID in male C57BL/6 wild-type and C3aR-knockout (C3aR-/-) mice. Cerebral blood flow (CBF) changes, hippocampal atrophy (HA), white matter degeneration (WMD), and ventricular size were assessed at 4 months post-BCAS using laser speckle contrast analysis (LSCI) and magnetic resonance imaging (MRI). Cognitive function was evaluated using the Morris water maze (MWM), and novel object recognition (NOR), immunostaining, and western blot were performed to assess the effect of genetic C3aR deletion on post-VCID outcomes. BCAS resulted in decreased CBF and increased HA, WMD, and neurovascular inflammation in WT (C57BL/6) compared to C3aR-/- (C3aR-KO) mice. Moreover, C3aR-/- mice exhibited improved cognitive function on NOR and MWM relative to WT controls. We conclude that over-activation of the C3a/C3aR axis exacerbates neurovascular inflammation leading to poor VCID outcomes which are mitigated by C3aR deletion. Future studies are warranted to dissect the role of cell-specific C3aR in VCID.
Collapse
Affiliation(s)
- Kanchan Bhatia
- Department of Neurosurgery, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA
- School of Mathematical and Natural Sciences, Arizona State University, Phoenix, AZ, USA
| | - Adam Kindelin
- Department of Neurosurgery, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA
| | - Muhammad Nadeem
- Department of Translational Neuroscience, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA
| | | | - Junxiang Yin
- Department of Translational Neuroscience, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA
- Department of Neurology, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA
| | - Alberto Fuentes
- Barrow Neurological Institute/Arizona State University Center for Preclinical Imaging, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA
| | - Karis Miller
- Department of Neurosurgery, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA
- Department of Translational Neuroscience, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA
| | - Gregory H Turner
- Barrow Neurological Institute/Arizona State University Center for Preclinical Imaging, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA
| | - Mark C Preul
- Department of Neurosurgery, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA
| | - Abdullah S Ahmad
- Department of Translational Neuroscience, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA
- Department of Neurology, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA
| | - Elliott J Mufson
- Department of Translational Neuroscience, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA
| | - Michael F Waters
- Department of Translational Neuroscience, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA
- Department of Neurology, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA
| | - Saif Ahmad
- Department of Neurosurgery, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA.
- Department of Translational Neuroscience, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85013, USA.
| | - Andrew F Ducruet
- Departments of Neurosurgery & Translational Neuroscience, Barrow Neurological Institute, SJHMC, Dignity Health, Phoenix, AZ, 85086, USA.
| |
Collapse
|
35
|
Elman-Shina K, Efrati S. Ischemia as a common trigger for Alzheimer’s disease. Front Aging Neurosci 2022; 14:1012779. [PMID: 36225888 PMCID: PMC9549288 DOI: 10.3389/fnagi.2022.1012779] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 08/29/2022] [Indexed: 11/15/2022] Open
Abstract
Alzheimer’s disease has various potential etiologies, all culminating in the accumulation of beta -amyloid derivatives and significant cognitive decline. Vascular-related pathology is one of the more frequent etiologies, especially in persons older than 65 years, as vascular risk factors are linked to both cerebrovascular disease and the development of AD. The vascular patho-mechanism includes atherosclerosis, large and small vessel arteriosclerosis, cortical and subcortical infarcts, white matter lesions, and microbleeds. These insults cause hypoperfusion, tissue ischemia, chronic inflammation, neuronal death, gliosis, cerebral atrophy, and accumulation of beta-amyloid and phosphorylated tau proteins. In preclinical studies, hyperbaric oxygen therapy has been shown to reverse brain ischemia, and thus alleviate inflammation, reverse the accumulation of beta-amyloid, induce regeneration of axonal white matter, stimulate axonal growth, promote blood–brain barrier integrity, reduce inflammatory reactions, and improve brain performance. In this perspective article we will summarize the patho-mechanisms induced by brain ischemia and their contribution to the development of AD. We will also review the potential role of interventions that aim to reverse brain ischemia, and discuss their relevance for clinical practice.
Collapse
Affiliation(s)
- Karin Elman-Shina
- Sagol Center for Hyperbaric Medicine and Research, Shamir Medical Center (Assaf Harofeh), Tzerifin, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- *Correspondence: Karin Elman-Shina,
| | - Shai Efrati
- Sagol Center for Hyperbaric Medicine and Research, Shamir Medical Center (Assaf Harofeh), Tzerifin, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- Research and Development Unit, Shamir Medical Center (Assaf Harofeh), Tzerifin, Israel
| |
Collapse
|
36
|
Ligustilide Improves Cognitive Impairment via Regulating the SIRT1/IRE1α/XBP1s/CHOP Pathway in Vascular Dementia Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:6664990. [PMID: 36017237 PMCID: PMC9398841 DOI: 10.1155/2022/6664990] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 07/20/2022] [Accepted: 07/29/2022] [Indexed: 11/24/2022]
Abstract
Vascular dementia (VaD), the second cause of dementia, is caused by chronic cerebral hypoperfusion, producing progressive damage to cerebral cortex, hippocampus, and white matter. Ligustilide (LIG), one of the main active ingredients of Angelica sinensis, exerts the neuroprotective effect on neurodegenerative diseases. However, the mechanism remains unclear. An in vivo model of bilateral common carotid artery occlusion and in vitro model of oxygen glucose deprivation (OGD) were employed in this study. LIG (20 or 40 mg/kg/day) was intragastrically administered to the VaD rats for four weeks. The results of the Morris water maze test demonstrated that LIG effectively ameliorated learning and memory deficiency in VaD rats. LIG obviously relieved neuronal oxidative stress damage by increasing the activities of catalase (CAT), superoxide dismutase (SOD), and glutathione peroxidase (GSH-PX) and decreasing the level of malondialdehyde (MDA) in VaD rats. Nissl staining showed that LIG increased the number of the Nissl body in VaD rats. After LIG administration, the apoptotic-related protein, Bax, was decreased and Bcl-2 was increased in the hippocampus of VaD rats. Moreover, the expressions of sirtuin 1 (SIRT1) and protein disulfide isomerase (PDI) were decreased, binding immunoglobulin protein (BIP) and phospho-inositol-requiring enzyme-1α (P-IRE1α), X-box binding protein 1 (XBP1s), and C/EBP-homologous protein (CHOP) were increased in VaD rats. After LIG treatment, these changes were reversed. The immunofluorescence results further showed that LIG upregulated the expression of SIRT1 and downregulated the expression of P-IRE1α in VaD rats. In addition, in vitro experiment showed that EX-527 (SIRT1 inhibitor) partly abolished the inhibitory effect of LIG on the IRE1α/XBP1s/CHOP pathway. In conclusion, these studies indicated that LIG could improve cognitive impairment by regulating the SIRT1/IRE1α/XBP1s/CHOP pathway in VaD rats.
Collapse
|
37
|
Yu W, Li Y, Hu J, Wu J, Huang Y. A Study on the Pathogenesis of Vascular Cognitive Impairment and Dementia: The Chronic Cerebral Hypoperfusion Hypothesis. J Clin Med 2022; 11:jcm11164742. [PMID: 36012981 PMCID: PMC9409771 DOI: 10.3390/jcm11164742] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/27/2022] [Accepted: 08/02/2022] [Indexed: 11/16/2022] Open
Abstract
The pathogenic mechanisms underlying vascular cognitive impairment and dementia (VCID) remain controversial due to the heterogeneity of vascular causes and complexity of disease neuropathology. However, one common feature shared among all these vascular causes is cerebral blood flow (CBF) dysregulation, and chronic cerebral hypoperfusion (CCH) is the universal consequence of CBF dysregulation, which subsequently results in an insufficient blood supply to the brain, ultimately contributing to VCID. The purpose of this comprehensive review is to emphasize the important contributions of CCH to VCID and illustrate the current findings about the mechanisms involved in CCH-induced VCID pathological changes. Specifically, evidence is mainly provided to support the molecular mechanisms, including Aβ accumulation, inflammation, oxidative stress, blood-brain barrier (BBB) disruption, trophic uncoupling and white matter lesions (WMLs). Notably, there are close interactions among these multiple mechanisms, and further research is necessary to elucidate the hitherto unsolved questions regarding these interactions. An enhanced understanding of the pathological features in preclinical models could provide a theoretical basis, ultimately achieving the shift from treatment to prevention.
Collapse
Affiliation(s)
- Weiwei Yu
- Department of Neurology, Peking University Shenzhen Hospital, 1120 Lianhua Road, Futian District, Shenzhen 518036, China
| | - Yao Li
- Department of Neurology, Peking University Shenzhen Hospital, 1120 Lianhua Road, Futian District, Shenzhen 518036, China
| | - Jun Hu
- Department of Neurology, Peking University Shenzhen Hospital, 1120 Lianhua Road, Futian District, Shenzhen 518036, China
| | - Jun Wu
- Department of Neurology, Peking University Shenzhen Hospital, 1120 Lianhua Road, Futian District, Shenzhen 518036, China
- Correspondence: (J.W.); (Y.H.); Tel.: +86-0755-8392-2833 (J.W.); +86-010-83572857 (Y.H.)
| | - Yining Huang
- Department of Neurology, Peking University First Hospital, 8 Xishiku Street Xicheng District, Beijing 100034, China
- Correspondence: (J.W.); (Y.H.); Tel.: +86-0755-8392-2833 (J.W.); +86-010-83572857 (Y.H.)
| |
Collapse
|
38
|
Nakase T, Tatewaki Y, Thyreau B, Mutoh T, Tomita N, Yamamoto S, Takano Y, Muranaka M, Taki Y. Impact of constipation on progression of Alzheimer's disease: A retrospective study. CNS Neurosci Ther 2022; 28:1964-1973. [PMID: 35934956 PMCID: PMC9627372 DOI: 10.1111/cns.13940] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 07/26/2022] [Accepted: 07/27/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND AND PURPOSE In terms of the gut-brain axis, constipation has been considered to be an important factor of neurodegenerative diseases, although the exact mechanism is still controversial. Herein, we aimed to investigate the contribution of constipation to the progression of dementia in a retrospective study. METHODS Patients of Alzheimer's disease(AD) and amnestic mild cognitive impairment were consecutively screened between January 2015 and December 2020, and those of whom brain MRI and neuropsychological tests were performed twice were enrolled in this study. Participants were classified into with constipation (Cons[+], n = 20) and without constipation (Cons[-], n = 64) groups. Laboratory data at the first visit were used. Regression analysis was performed in MMSE, ADAS-Cog, and the volumes of hippocampus on MRI-MPRAGE images and deep white matter lesions (DWMLs) on MRI-FLAIR images obtained at two different time points. RESULTS The main finding was that the Cons[+] group showed 2.7 times faster decline in cognitive impairment compared with the Cons[-] group, that is, the liner coefficients of ADAS-Cog were 2.3544 points/year in the Cons[+] and 0.8592 points/year in the Cons[-] groups. Ancillary, changes of DWMLs showed significant correlation with the time span (p < 0.01), and the liner coefficients of DWMLs were 24.48 ml/year in the Cons[+] and 14.83 ml/year in the Cons[-] group, although annual rate of hippocampal atrophy was not different between the two groups. Moreover, serum homocysteine level at baseline was significantly higher in the Cons[+] group than Cons[-] group (14.6 ± 6.4 and 11.5 ± 4.2 nmol/ml, respectively: p = 0.03). CONCLUSION There is a significant correlation between constipation and faster progression of AD symptoms along with expansion of DWMLs.
Collapse
Affiliation(s)
- Taizen Nakase
- Smart Aging Research CenterTohoku UniversitySendaiJapan,Department of Aging Research and Geriatric Medicine, Institute of Development, Aging and CancerTohoku UniversitySendaiJapan
| | - Yasuko Tatewaki
- Department of Aging Research and Geriatric Medicine, Institute of Development, Aging and CancerTohoku UniversitySendaiJapan
| | | | - Tatsushi Mutoh
- Department of Aging Research and Geriatric Medicine, Institute of Development, Aging and CancerTohoku UniversitySendaiJapan
| | - Naoki Tomita
- Department of Aging Research and Geriatric Medicine, Institute of Development, Aging and CancerTohoku UniversitySendaiJapan
| | - Shuzo Yamamoto
- Department of Aging Research and Geriatric Medicine, Institute of Development, Aging and CancerTohoku UniversitySendaiJapan
| | - Yumi Takano
- Department of Aging Research and Geriatric Medicine, Institute of Development, Aging and CancerTohoku UniversitySendaiJapan
| | - Michiho Muranaka
- Department of Aging Research and Geriatric Medicine, Institute of Development, Aging and CancerTohoku UniversitySendaiJapan
| | - Yasuyuki Taki
- Smart Aging Research CenterTohoku UniversitySendaiJapan,Department of Aging Research and Geriatric Medicine, Institute of Development, Aging and CancerTohoku UniversitySendaiJapan
| |
Collapse
|
39
|
Zhou Z, Ma Y, Xu T, Wu S, Yang GY, Ding J, Wang X. Deeper cerebral hypoperfusion leads to spatial cognitive impairment in mice. Stroke Vasc Neurol 2022; 7:527-533. [PMID: 35817499 PMCID: PMC9811541 DOI: 10.1136/svn-2022-001594] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 05/11/2022] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Vascular cognitive impairment (VCI) is the second-leading cause of dementia worldwide, which is caused by cerebrovascular diseases or relevant risk factors. However, there are no appropriate animal models, which can be used to study changes of neuropathology in the human VCI. To better understand the development of VCI, we modified three mouse models of chronical vascular diseases, and further compared the advantage and disadvantage of these models. We hope to establish a more suitable mouse model mimicking VCI in human beings. METHODS Adult male C57/BL6 mice (n=98) were used and animals underwent transient bilateral common carotid arteries occlusion (tBCCAO), or bilateral common carotid artery stenosis (BCAS), or right unilateral common carotid artery occlusion, respectively. Haemodynamic changes of surface cerebral blood flow (CBF) were examined up to 4 weeks. Spatial cognitive impairment was evaluated to determine the consequence of chronic cerebral ischaemia. RESULTS These mouse models showed different extents of CBF reduction and spatial reference memory impairment from 1 week up to 4 weeks postoperation compared with the control group (p<0.05). We found that (1) bilaterally ligation of common carotid artery caused decrease of 90% CBF in C57/BL6 mice (p<0.05) and caused acute instead of prolonged impairment of spatial reference memory (p<0.05); (2) unilateral ligation of common carotid artery did not cause severe ipsilateral ischaemia as seen in the tBCCAO mice and caused minor but significant spatial reference memory disturbance (p<0.05); and (3) 20% decrease in the bilateral CBF did not cause spatial reference memory impairment 4 weeks postoperation (p>0.05), while 30% decrease in bilateral or unilateral CBF led to significant memory disturbance in mice (p<0.05). CONCLUSION We demonstrated that BCAS using 0.16/0.18 mm microcoils is an alternative VCI mouse model when studying the mechanism and developing therapy of VCI.
Collapse
Affiliation(s)
- Zhiyuan Zhou
- Department of Neurology, Zhongshan Hospital Fudan University, Shanghai, China
| | - Yuanyuan Ma
- Department of Neurology, Zhongshan Hospital Fudan University, Shanghai, China
| | - Tongtong Xu
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Shengju Wu
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Guo-Yuan Yang
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Jing Ding
- Department of Neurology, Zhongshan Hospital Fudan University, Shanghai, China
| | - Xin Wang
- Department of Neurology, Zhongshan Hospital Fudan University, Shanghai, China,Department of the State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
40
|
Upregulation of PGC-1 α Attenuates Oxygen-Glucose Deprivation-Induced Hippocampal Neuronal Injury. Neural Plast 2022; 2022:9682999. [PMID: 35719138 PMCID: PMC9203239 DOI: 10.1155/2022/9682999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 05/25/2022] [Indexed: 11/30/2022] Open
Abstract
Hippocampal neuronal damage likely underlies cognitive impairment in vascular dementia (VaD). PPARγ coactivator-1α (PGC-1α) is a master regulator of mitochondrial biogenesis. However, the role and the precise mechanism of how PGC-1α alleviates hippocampal neuronal injury remain unknown. To address this question, HT-22 cells, an immortalized hippocampal neuron cell line, with or without PGC-1α overexpression were subjected to oxygen-glucose deprivation (OGD), which mimics the circumstance of chronic cerebral hypoperfusion in VaD. After OGD, cell viability was assessed using the MTS assay. The mitochondrial function and reactive oxygen species (ROS) were both detected. ChIP-Seq analysis was employed to discover the underlying molecular mechanism of PGC-1α-mediated neuroprotective effects. Our results showed that mitochondrial membrane potentials were increased and ROS production was decreased in PGC-1α overexpressing cells, which increased cell viability. The further bioinformatics analysis from ChIP-Seq data indicated that PGC-1α may participate in the regulation of apoptosis, autophagy, and mitophagy pathways in HT-22 cells. We found that PGC-1α promoted the LC3-II formation and reduced the neuronal apoptosis determined by TUNEL staining. In addition, PGC-1α upregulated the expressions of mitochondrial antioxidants, including SOD2, Trx2, and Prx3. In summary, our findings indicate that PGC-1α may attenuate OGD-induced hippocampal neuronal damage by regulating multiple mechanisms, like autophagy and mitochondrial function. Thus, PGC-1α may be a potential therapeutic target for hippocampal damage associated with cognitive impairment.
Collapse
|
41
|
Sun W, Huang L, Cheng Y, Qin R, Xu H, Shao P, Ma J, Yao Z, Shi L, Xu Y. Medial Temporal Atrophy Contributes to Cognitive Impairment in Cerebral Small Vessel Disease. Front Neurol 2022; 13:858171. [PMID: 35665031 PMCID: PMC9159509 DOI: 10.3389/fneur.2022.858171] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 04/14/2022] [Indexed: 11/18/2022] Open
Abstract
Background The role of brain atrophy in cognitive decline related to cerebral small vessel disease (CSVD) remains unclear. This study used AccuBrain™ to identify major CSVD-related brain changes and verified the relationship between brain atrophy and different cognition domains in CSVD patients. Methods All enrolled 242 CSVD patients and 76 healthy participants underwent magnetic resonance imaging examinations and detailed neuropsychological scale assessments were collected at the same time. The AccuBrain™ technology was applied to fully automated image segmentation, measurement, and calculation of the acquired imaging results to obtain the volumes of different brain partitions and the volume of WMH for quantitative analysis. Correlation analyses were used to estimate the relationship between MRI features and different cognitive domains. Multifactor linear regression models were performed to analyze independent predictors of MTA and cognitive decline. Results CSVD patients exhibited multiple gray matter nucleus volume decreases in the basal ganglia regions and brain lobes, including the temporal lobe (P = 0.019), especially in the medial temporal lobe (p < 0.001), parietal lobe (p = 0.013), and cingulate lobe (p = 0.036) compare to HC. The volume of PWMH was an independent predictor of MTA for CSVD patients. Both medial temporal atrophy (MTA) and PWMH were associated with cognition impairment in CSVD-CI patients. MTA mediated the effect of PWMH on executive function in CSVD-CI patients. Conclusions Our results showed that MTA was related to cognition impairment in CSVD patients, which might become a potential imaging marker for CSVD-CI.
Collapse
Affiliation(s)
- Wenshan Sun
- Department of Neurology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China
- Department of Neurology, Nanjing Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Jiangsu Key Laboratory for Molecular Medicine, Nanjing University, Nanjing, China
- Department of Neurology, Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Lili Huang
- Department of Neurology, Nanjing Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Jiangsu Key Laboratory for Molecular Medicine, Nanjing University, Nanjing, China
| | - Yue Cheng
- Department of Neurology, Nanjing Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Jiangsu Key Laboratory for Molecular Medicine, Nanjing University, Nanjing, China
| | - Ruomeng Qin
- Department of Neurology, Nanjing Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Jiangsu Key Laboratory for Molecular Medicine, Nanjing University, Nanjing, China
| | - Hengheng Xu
- Department of Neurology, Nanjing Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Jiangsu Key Laboratory for Molecular Medicine, Nanjing University, Nanjing, China
| | - Pengfei Shao
- Department of Neurology, Nanjing Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Jiangsu Key Laboratory for Molecular Medicine, Nanjing University, Nanjing, China
| | - Junyi Ma
- Department of Neurology, Nanjing Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Jiangsu Key Laboratory for Molecular Medicine, Nanjing University, Nanjing, China
| | - Zhelv Yao
- Department of Neurology, Nanjing Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Jiangsu Key Laboratory for Molecular Medicine, Nanjing University, Nanjing, China
| | - Lin Shi
- Department of Imaging and Interventional Radiology, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- BrainNow Research Institute, Hong Kong Science and Technology Park, Hong Kong, Hong Kong SAR, China
| | - Yun Xu
- Department of Neurology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China
- Department of Neurology, Nanjing Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Jiangsu Key Laboratory for Molecular Medicine, Nanjing University, Nanjing, China
- *Correspondence: Yun Xu
| |
Collapse
|
42
|
Lee JI, Lim JS, Hong JH, Kim S, Lee SW, Ji HD, Won KS, Song BI, Kim HW. Selective neurodegeneration of the hippocampus caused by chronic cerebral hypoperfusion: F-18 FDG PET study in rats. PLoS One 2022; 17:e0262224. [PMID: 35143502 PMCID: PMC8830734 DOI: 10.1371/journal.pone.0262224] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 12/20/2021] [Indexed: 11/20/2022] Open
Abstract
Background Chronic cerebral hypoperfusion (CCH) is known to induce Alzheimer’s disease (AD) pathology, but its mechanism remains unclear. The purpose of this study was to identify the cerebral regions that are affected by CCH, and to evaluate the development of AD pathology in a rat model of CCH. Methods A rat model of CCH was established by bilaterally ligating the common carotid arteries in adult male rats (CCH group). The identical operations were performed on sham rats without arteries ligation (control group). Regional cerebral glucose metabolism was evaluated at 1 and 3 months after bilateral CCA ligation using positron emission tomography with F-18 fluorodeoxyglucose. The expression levels of amyloid β40 (Aβ40), amyloid β42 (Aβ42), and hyperphosphorylated tau were evaluated using western blots at 3 months after the ligation. Cognitive function was evaluated using the Y-maze test at 3 months after the ligation. Results At 1 month after the ligation, cerebral glucose metabolism in the entorhinal, frontal association, motor, and somatosensory cortices were significantly decreased in the CCH group compared with those in the control group. At 3 months after the ligation, cerebral glucose metabolism was normalized in all regions except for the anterodorsal hippocampus, which was significantly decreased compared with that of the control group. The expression of Aβ42 and the Aβ42/40 ratio were significantly higher in the CCH group than those in the control group. The phosphorylated-tau levels of the hippocampus in the CCH group were significantly lower than those in the control group. Cognitive function was more impaired in the CCH group than that in the control group. Conclusion Our findings suggest that CCH causes selective neurodegeneration of the anterodorsal hippocampus, which may be a trigger point for the development of AD pathology.
Collapse
Affiliation(s)
- Jung-In Lee
- Department of Nuclear Medicine, Keimyung University Dongsan Hospital, Daegu, Republic of Korea
| | - Ji Sun Lim
- Department of Nuclear Medicine, Keimyung University Dongsan Hospital, Daegu, Republic of Korea
| | - Jeong-Ho Hong
- Department of Neurology, Keimyung University Dongsan Hospital, Daegu, Republic of Korea
| | - Shin Kim
- Department of Immunology, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Sang-Woo Lee
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Hyun Dong Ji
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Kyoung Sook Won
- Department of Nuclear Medicine, Keimyung University Dongsan Hospital, Daegu, Republic of Korea
| | - Bong-Il Song
- Department of Nuclear Medicine, Keimyung University Dongsan Hospital, Daegu, Republic of Korea
| | - Hae Won Kim
- Department of Nuclear Medicine, Keimyung University Dongsan Hospital, Daegu, Republic of Korea
- Department of Nuclear Medicine, School of Medicine & Institute for Medical Science, Keimyung University, Daegu, Korea
- * E-mail:
| |
Collapse
|
43
|
Hall GR, Boehm-Sturm P, Dirnagl U, Finke C, Foddis M, Harms C, Koch SP, Kuchling J, Madan CR, Mueller S, Sassi C, Sotiropoulos SN, Trueman RC, Wallis MD, Yildirim F, Farr TD. Long-Term Connectome Analysis Reveals Reshaping of Visual, Spatial Networks in a Model With Vascular Dementia Features. Stroke 2022; 53:1735-1745. [PMID: 35105183 PMCID: PMC9022688 DOI: 10.1161/strokeaha.121.036997] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Connectome analysis of neuroimaging data is a rapidly expanding field that offers the potential to diagnose, characterize, and predict neurological disease. Animal models provide insight into biological mechanisms that underpin disease, but connectivity approaches are currently lagging in the rodent.
Collapse
Affiliation(s)
- Gerard R Hall
- School of Life Sciences, University of Nottingham, United Kingdom (G.R.H., R.C.T., M.D.W., T.D.F.)
| | - Philipp Boehm-Sturm
- Department of Experimental Neurology, Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin (P.B.-S., U.D., M.F., C.H., S.P.K., S.M., C.S., T.D.F.).,corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Germany. NeuroCure Cluster of Excellence and Charité Core Facility 7T Experimental MRIs, Charité-Universitätsmedizin Berlin, Germany. (P.B.-S., U.D., M.F., C.H., S.P.K., S.M., C.S., F.Y., T.D.F.)
| | - Ulrich Dirnagl
- Department of Experimental Neurology, Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin (P.B.-S., U.D., M.F., C.H., S.P.K., S.M., C.S., T.D.F.).,corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Germany. NeuroCure Cluster of Excellence and Charité Core Facility 7T Experimental MRIs, Charité-Universitätsmedizin Berlin, Germany. (P.B.-S., U.D., M.F., C.H., S.P.K., S.M., C.S., F.Y., T.D.F.).,German Center for Neurodegenerative Diseases, Berlin Site, Germany (U.D.)
| | - Carsten Finke
- Department of Neurology, Charité-Universitätsmedizin Berlin, Germany. (C.F., J.K.).,Berlin School of Mind and Brain, Humboldt Universität zu Berlin, Germany (C.F.)
| | - Marco Foddis
- Department of Experimental Neurology, Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin (P.B.-S., U.D., M.F., C.H., S.P.K., S.M., C.S., T.D.F.).,corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Germany. NeuroCure Cluster of Excellence and Charité Core Facility 7T Experimental MRIs, Charité-Universitätsmedizin Berlin, Germany. (P.B.-S., U.D., M.F., C.H., S.P.K., S.M., C.S., F.Y., T.D.F.)
| | - Christoph Harms
- Department of Experimental Neurology, Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin (P.B.-S., U.D., M.F., C.H., S.P.K., S.M., C.S., T.D.F.).,corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Germany. NeuroCure Cluster of Excellence and Charité Core Facility 7T Experimental MRIs, Charité-Universitätsmedizin Berlin, Germany. (P.B.-S., U.D., M.F., C.H., S.P.K., S.M., C.S., F.Y., T.D.F.)
| | - Stefan Paul Koch
- Department of Experimental Neurology, Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin (P.B.-S., U.D., M.F., C.H., S.P.K., S.M., C.S., T.D.F.).,corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Germany. NeuroCure Cluster of Excellence and Charité Core Facility 7T Experimental MRIs, Charité-Universitätsmedizin Berlin, Germany. (P.B.-S., U.D., M.F., C.H., S.P.K., S.M., C.S., F.Y., T.D.F.)
| | - Joseph Kuchling
- Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité-Universitätsmedizin Berlin (J.K.).,Department of Neurology, Charité-Universitätsmedizin Berlin, Germany. (C.F., J.K.)
| | | | - Susanne Mueller
- Department of Experimental Neurology, Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin (P.B.-S., U.D., M.F., C.H., S.P.K., S.M., C.S., T.D.F.).,corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Germany. NeuroCure Cluster of Excellence and Charité Core Facility 7T Experimental MRIs, Charité-Universitätsmedizin Berlin, Germany. (P.B.-S., U.D., M.F., C.H., S.P.K., S.M., C.S., F.Y., T.D.F.)
| | - Celeste Sassi
- Department of Experimental Neurology, Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin (P.B.-S., U.D., M.F., C.H., S.P.K., S.M., C.S., T.D.F.).,corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Germany. NeuroCure Cluster of Excellence and Charité Core Facility 7T Experimental MRIs, Charité-Universitätsmedizin Berlin, Germany. (P.B.-S., U.D., M.F., C.H., S.P.K., S.M., C.S., F.Y., T.D.F.)
| | - Stamatios N Sotiropoulos
- Sir Peter Mansfield Imaging Centre, School of Medicine, University of Nottingham, United Kingdom (S.N.S.).,Centre for Functional MRI of the Brain, University of Oxford, United Kingdom (S.N.S.)
| | - Rebecca C Trueman
- School of Life Sciences, University of Nottingham, United Kingdom (G.R.H., R.C.T., M.D.W., T.D.F.)
| | - Marcus D Wallis
- School of Life Sciences, University of Nottingham, United Kingdom (G.R.H., R.C.T., M.D.W., T.D.F.)
| | - Ferah Yildirim
- corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Germany. NeuroCure Cluster of Excellence and Charité Core Facility 7T Experimental MRIs, Charité-Universitätsmedizin Berlin, Germany. (P.B.-S., U.D., M.F., C.H., S.P.K., S.M., C.S., F.Y., T.D.F.).,NeuroCure Cluster of Excellence and Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, Germany. (F.Y.)
| | - Tracy D Farr
- School of Life Sciences, University of Nottingham, United Kingdom (G.R.H., R.C.T., M.D.W., T.D.F.).,Department of Experimental Neurology, Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin (P.B.-S., U.D., M.F., C.H., S.P.K., S.M., C.S., T.D.F.).,corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Germany. NeuroCure Cluster of Excellence and Charité Core Facility 7T Experimental MRIs, Charité-Universitätsmedizin Berlin, Germany. (P.B.-S., U.D., M.F., C.H., S.P.K., S.M., C.S., F.Y., T.D.F.)
| |
Collapse
|
44
|
Mun J, Jung J, Park C. Effects of cerebral hypoperfusion on the cerebral white matter: a meta‑analysis. Acta Neurobiol Exp (Wars) 2021; 81:295-306. [PMID: 34672300 DOI: 10.21307/ane-2021-029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Decreased cerebral blood flow (CBF) in aging is known to induce aging‑related cerebral deteriorations, such as neuronal degeneration, white matter (WM) alterations, and vascular deformations. However, the effects of cerebral hypoperfusion on WM alterations remain unclear. This study investigates the relationship between cerebral hypoperfusion and WM total volume changes by assessing the trends in CBF and WM changes by meta‑analysis. In this meta‑analysis, the differences in CBF were compared according to cerebral hypoperfusion type and the effect of cerebral hypoperfusion on the total volume of WM changes in rodents. Using subgroup analysis, 13 studies were evaluated for comparing CBF according to the type of cerebral hypoperfusion; 12 studies were evaluated for comparing the effects of cerebral hypoperfusion on the total volume of WM changes. Our meta‑analysis shows that the total volume of WM decreases with a decrease in CBF. However, the reduction in\r\nthe total volume of WM was greater in normal aging mice than in the cerebral hypoperfusion model mice. These results suggest that the reduction of cerebral WM volume during the aging process is affected by other factors in addition to a decrease in CBF.
Collapse
Affiliation(s)
- Juyeon Mun
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea
- Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Junyang Jung
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea
- Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Chan Park
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea
- Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul, Republic of Korea;
| |
Collapse
|
45
|
Fan Y, Shen M, Huo Y, Gao X, Li C, Zheng R, Zhang J. Total Cerebral Small Vessel Disease Burden on MRI Correlates With Medial Temporal Lobe Atrophy and Cognitive Performance in Patients of a Memory Clinic. Front Aging Neurosci 2021; 13:698035. [PMID: 34566621 PMCID: PMC8456168 DOI: 10.3389/fnagi.2021.698035] [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: 04/20/2021] [Accepted: 08/09/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Cerebral small vessel disease (cSVD) and neurodegeneration are the two main causes of dementia and are considered distinct pathological processes, while studies have shown overlaps and interactions between the two pathological pathways. Medial temporal atrophy (MTA) is considered a classic marker of neurodegeneration. We aimed to investigate the relationship of total cSVD burden and MTA on MRI using a total cSVD score and to explore the impact of the two MRI features on cognition. Methods: Patients in a memory clinic were enrolled, who underwent brain MRI scan and cognitive evaluation within 7 days after the first visit. MTA and total cSVD score were rated using validated visual scales. Cognitive function was assessed by using Mini-Mental State Examination (MMSE) and Montreal Cognitive Assessment (MoCA) scales. Spearman's correlation and regression models were used to test (i) the association between MTA and total cSVD score as well as each cSVD marker and (ii) the correlation of the MRI features and cognitive status. Results: A total of 312 patients were finally enrolled, with a median age of 75.0 (66.0-80.0) years and 40.7% (127/312) males. All of them finished MRI and MMSE, and 293 subjects finished MoCA. Of note, 71.8% (224/312) of the patients had at least one of the cSVD markers, and 48.7% (152/312) of them had moderate-severe MTA. The total cSVD score was independently associated with MTA levels, after adjusting for age, gender, years of education, and other vascular risk factors (OR 1.191, 95% CI 1.071-1.324, P = 0.001). In regard to individual markers, a significant association existed only between white matter hyperintensities and MTA after adjusting for the factors mentioned above (OR 1.338, 95% CI 1.050-1.704, P = 0.018). Both MTA and total cSVD score were independent risk factors for MMSE ≤ 26 (MTA: OR 1.877, 95% CI 1.407-2.503, P < 0.001; total cSVD score: OR 1.474, 95% CI 1.132-1.921, P = 0.004), and MoCA < 26 (MTA: OR 1.629, 95% CI 1.112-2.388, P = 0.012; total cSVD score: OR 1.520, 95% CI 1.068-2.162, P = 0.020). Among all the cSVD markers, microbleed was found significantly associated with MMSE ≤ 26, while no marker was demonstrated a relationship with MoCA < 26. Conclusion: Cerebral small vessel disease was related to MTA in patients of a memory clinic, and both the MRI features had a significant association with cognitive impairment.
Collapse
Affiliation(s)
- Yangyi Fan
- Department of Neurology, Peking University People's Hospital, Beijing, China
| | - Ming Shen
- Department of Neurology, Peking University People's Hospital, Beijing, China
| | - Yang Huo
- Department of Neurology, Peking University People's Hospital, Beijing, China
| | - Xuguang Gao
- Department of Neurology, Peking University People's Hospital, Beijing, China
| | - Chun Li
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, China
| | - Ruimao Zheng
- Neuroscience Research Institute, Peking University, Beijing, China
| | - Jun Zhang
- Department of Neurology, Peking University People's Hospital, Beijing, China
| |
Collapse
|
46
|
Poh L, Fann DY, Wong P, Lim HM, Foo SL, Kang SW, Rajeev V, Selvaraji S, Iyer VR, Parathy N, Khan MB, Hess DC, Jo DG, Drummond GR, Sobey CG, Lai MKP, Chen CLH, Lim LHK, Arumugam TV. AIM2 inflammasome mediates hallmark neuropathological alterations and cognitive impairment in a mouse model of vascular dementia. Mol Psychiatry 2021; 26:4544-4560. [PMID: 33299135 DOI: 10.1038/s41380-020-00971-5] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 11/12/2020] [Accepted: 11/23/2020] [Indexed: 12/11/2022]
Abstract
Chronic cerebral hypoperfusion is associated with vascular dementia (VaD). Cerebral hypoperfusion may initiate complex molecular and cellular inflammatory pathways that contribute to long-term cognitive impairment and memory loss. Here we used a bilateral common carotid artery stenosis (BCAS) mouse model of VaD to investigate its effect on the innate immune response-particularly the inflammasome signaling pathway. Comprehensive analyses revealed that chronic cerebral hypoperfusion induces a complex temporal expression and activation of inflammasome components and their downstream products (IL-1β and IL-18) in different brain regions, and promotes activation of apoptotic and pyroptotic cell death pathways. Polarized glial-cell activation, white-matter lesion formation and hippocampal neuronal loss also occurred in a spatiotemporal manner. Moreover, in AIM2 knockout mice we observed attenuated inflammasome-mediated production of proinflammatory cytokines, apoptosis, and pyroptosis, as well as resistance to chronic microglial activation, myelin breakdown, hippocampal neuronal loss, and behavioral and cognitive deficits following BCAS. Hence, we have demonstrated that activation of the AIM2 inflammasome substantially contributes to the pathophysiology of chronic cerebral hypoperfusion-induced brain injury and may therefore represent a promising therapeutic target for attenuating cognitive impairment in VaD.
Collapse
Affiliation(s)
- Luting Poh
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - David Y Fann
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore. .,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | - Peiyan Wong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Neuroscience and Behavioural Disorders Programme, Duke-NUS Medical School, Singapore, Singapore
| | - Hong Meng Lim
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Sok Lin Foo
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Sung-Wook Kang
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Vismitha Rajeev
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Sharmelee Selvaraji
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Vinaya Rajagopal Iyer
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Nageiswari Parathy
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | | | - David C Hess
- Department of Neurology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Grant R Drummond
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, Australia
| | - Christopher G Sobey
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, Australia
| | - Mitchell K P Lai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Memory, Aging and Cognition Centre, National University Health System, Singapore, Singapore
| | - Christopher Li-Hsian Chen
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Memory, Aging and Cognition Centre, National University Health System, Singapore, Singapore
| | - Lina H K Lim
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Thiruma V Arumugam
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore. .,School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea. .,Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC, Australia.
| |
Collapse
|
47
|
Yildirim F, Foddis M, Blumenau S, Müller S, Kajetan B, Holtgrewe M, Kola V, Beule D, Sassi C. Shared and oppositely regulated transcriptomic signatures in Huntington's disease and brain ischemia confirm known and unveil novel potential neuroprotective genes. Neurobiol Aging 2021; 104:122.e1-122.e17. [PMID: 33875290 DOI: 10.1016/j.neurobiolaging.2021.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 02/13/2021] [Accepted: 03/02/2021] [Indexed: 11/20/2022]
Abstract
Huntington's disease and subcortical vascular dementia display similar dementing features, shaped by different degrees of striatal atrophy, deep white matter degeneration and tau pathology. To investigate the hypothesis that Huntington's disease transcriptomic hallmarks may provide a window into potential protective genes upregulated during brain acute and subacute ischemia, we compared RNA sequencing signatures in the most affected brain areas of 2 widely used experimental mouse models: Huntington's disease, (R6/2, striatum and cortex and Q175, hippocampus) and brain ischemia-subcortical vascular dementia (BCCAS, striatum, cortex and hippocampus). We identified a cluster of 55 shared genes significantly differentially regulated in both models and we screened these in 2 different mouse models of Alzheimer's disease, and 96 early-onset familial and apparently sporadic small vessel ischemic disease patients. Our data support the prevalent role of transcriptional regulation upon genetic coding variability of known neuroprotective genes (Egr2, Fos, Ptgs2, Itga5, Cdkn1a, Gsn, Npas4, Btg2, Cebpb) and provide a list of potential additional ones likely implicated in different dementing disorders and worth further investigation.
Collapse
Affiliation(s)
- Ferah Yildirim
- Department of Neuropsychiatry, Department of Psychiatry and Psychotherapy, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Marco Foddis
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Sonja Blumenau
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Susanne Müller
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Bentele Kajetan
- Berlin Institute of Health, BIH, Core Unit Bioinformatics, Berlin, Germany
| | - Manuel Holtgrewe
- Berlin Institute of Health, BIH, Core Unit Bioinformatics, Berlin, Germany
| | - Vasilis Kola
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Dieter Beule
- Berlin Institute of Health, BIH, Core Unit Bioinformatics, Berlin, Germany
| | - Celeste Sassi
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.
| |
Collapse
|
48
|
Youssef MI, Ma J, Chen Z, Hu WW. Potential therapeutic agents for ischemic white matter damage. Neurochem Int 2021; 149:105116. [PMID: 34229025 DOI: 10.1016/j.neuint.2021.105116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 06/24/2021] [Indexed: 11/19/2022]
Abstract
Ischemic white matter damage (WMD) is increasingly being considered as one of the major causes of neurological disorders in older adults and preterm infants. The functional consequences of WMD triggers a progressive cognitive decline and dementia particularly in patients with ischemic cerebrovascular diseases. Despite the major stride made in the pathogenesis mechanisms of ischemic WMD in the last century, effective medications are still not available. So, there is an urgent need to explore a promising approach to slow the progression or modify its pathological course. In this review, we discussed the animal models, the pathological mechanisms and the potential therapeutic agents for ischemic WMD. The development in the studies of anti-oxidants, free radical scavengers, anti-inflammatory or anti-apoptotic agents and neurotrophic factors in ischemic WMD were summarized. The agents which either alleviate oligodendrocyte damage or promote its proliferation or differentiation may have potential value for the treatment of ischemic WMD. Moreover, drugs with multifaceted protective activities or a wide therapeutic window may be optimal for clinical translation.
Collapse
Affiliation(s)
- Mahmoud I Youssef
- Department of Pharmacology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, PR China
| | - Jing Ma
- Department of Pharmacy, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, PR China.
| | - Zhong Chen
- Department of Pharmacology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, PR China; Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, PR China.
| | - Wei-Wei Hu
- Department of Pharmacology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, PR China.
| |
Collapse
|
49
|
An L, Chopp M, Zacharek A, Shen Y, Chen Z, Qian Y, Li W, Landschoot-Ward J, Liu Z, Venkat P. Cardiac Dysfunction in a Mouse Vascular Dementia Model of Bilateral Common Carotid Artery Stenosis. Front Cardiovasc Med 2021; 8:681572. [PMID: 34179145 PMCID: PMC8225957 DOI: 10.3389/fcvm.2021.681572] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 05/12/2021] [Indexed: 12/29/2022] Open
Abstract
Background: Cardiac function is associated with cognitive function. Previously, we found that stroke and traumatic brain injury evoke cardiac dysfunction in mice. In this study, we investigate whether bilateral common carotid artery stenosis (BCAS), a model that induces vascular dementia (VaD) in mice, induces cardiac dysfunction. Methods: Late-adult (6-8 months) C57BL/6J mice were subjected to sham surgery (n = 6) or BCAS (n = 8). BCAS was performed by applying microcoils (0.16 mm internal diameter) around both common carotid arteries. Cerebral blood flow and cognitive function tests were performed 21-28 days post-BCAS. Echocardiography was conducted in conscious mice 29 days after BCAS. Mice were sacrificed 30 days after BCAS. Heart tissues were isolated for immunohistochemical evaluation and real-time PCR assay. Results: Compared to sham mice, BCAS in mice significantly induced cerebral hypoperfusion and cognitive dysfunction, increased cardiac hypertrophy, as indicated by the increased heart weight and the ratio of heart weight/body weight, and induced cardiac dysfunction and left ventricular (LV) enlargement, indicated by a decreased LV ejection fraction (LVEF) and LV fractional shortening (LVFS), increased LV dimension (LVD), and increased LV mass. Cognitive deficits significantly correlated with cardiac deficits. BCAS mice also exhibited significantly increased cardiac fibrosis, increased oxidative stress, as indicated by 4-hydroxynonenal and NADPH oxidase-2, increased leukocyte and macrophage infiltration into the heart, and increased cardiac interleukin-6 and thrombin gene expression. Conclusions: BCAS in mice without primary cardiac disease provokes cardiac dysfunction, which, in part, may be mediated by increased inflammation and oxidative stress.
Collapse
Affiliation(s)
- Lulu An
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States.,Department of Physics, Oakland University, Rochester, MI, United States
| | - Alex Zacharek
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Yi Shen
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Zhili Chen
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Yu Qian
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Wei Li
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | | | - Zhongwu Liu
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| | - Poornima Venkat
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
| |
Collapse
|
50
|
Kim KJ, Diaz JR, Presa JL, Muller PR, Brands MW, Khan MB, Hess DC, Althammer F, Stern JE, Filosa JA. Decreased parenchymal arteriolar tone uncouples vessel-to-neuronal communication in a mouse model of vascular cognitive impairment. GeroScience 2021; 43:1405-1422. [PMID: 33410092 PMCID: PMC8190257 DOI: 10.1007/s11357-020-00305-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 11/22/2020] [Indexed: 01/18/2023] Open
Abstract
Chronic hypoperfusion is a key contributor to cognitive decline and neurodegenerative conditions, but the cellular mechanisms remain ill-defined. Using a multidisciplinary approach, we sought to elucidate chronic hypoperfusion-evoked functional changes at the neurovascular unit. We used bilateral common carotid artery stenosis (BCAS), a well-established model of vascular cognitive impairment, combined with an ex vivo preparation that allows pressurization of parenchymal arterioles in a brain slice. Our results demonstrate that mild (~ 30%), chronic hypoperfusion significantly altered the functional integrity of the cortical neurovascular unit. Although pial cerebral perfusion recovered over time, parenchymal arterioles progressively lost tone, exhibiting significant reductions by day 28 post-surgery. We provide supportive evidence for reduced adenosine 1 receptor-mediated vasoconstriction as a potential mechanism in the adaptive response underlying the reduced baseline tone in parenchymal arterioles. In addition, we show that in response to the neuromodulator adenosine, the action potential frequency of cortical pyramidal neurons was significantly reduced in all groups. However, a significant decrease in adenosine-induced hyperpolarization was observed in BCAS 14 days. At the microvascular level, constriction-induced inhibition of pyramidal neurons was significantly compromised in BCAS mice. Collectively, these results suggest that BCAS uncouples vessel-to-neuron communication-vasculo-neuronal coupling-a potential early event in cognitive decline.
Collapse
Affiliation(s)
- Ki Jung Kim
- Department of Physiology, Augusta University, Augusta, GA, 30912, USA
| | - Juan Ramiro Diaz
- Department of Physiology, Augusta University, Augusta, GA, 30912, USA
| | - Jessica L Presa
- Department of Physiology, Augusta University, Augusta, GA, 30912, USA
| | - P Robinson Muller
- Department of Physiology, Augusta University, Augusta, GA, 30912, USA
| | - Michael W Brands
- Department of Physiology, Augusta University, Augusta, GA, 30912, USA
| | - Mohammad B Khan
- Department of Neurology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - David C Hess
- Department of Neurology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | | | - Javier E Stern
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA
| | - Jessica A Filosa
- Department of Physiology, Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|