1
|
Anderson ME, Wind EJ, Robison LS. Exploring the neuroprotective role of physical activity in cerebral small vessel disease. Brain Res 2024; 1833:148884. [PMID: 38527712 DOI: 10.1016/j.brainres.2024.148884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 03/19/2024] [Accepted: 03/20/2024] [Indexed: 03/27/2024]
Abstract
Cerebral small vessel disease (cSVD) is a common neurological finding characterized by abnormalities of the small blood vessels in the brain. Previous research has established a strong connection between cSVD and stroke, as well as neurodegenerative disorders, notably Alzheimer's disease (AD) and other dementias. As the search for effective interventions continues, physical activity (PA) has emerged as a potential preventative and therapeutic avenue. This review synthesizes the human and animal literature on the influence of PA on cSVD, highlighting the importance of determining optimal exercise protocols, considering aspects such as intensity, duration, timing, and exercise type. Furthermore, the necessity of widening the age bracket in research samples is discussed, ensuring a holistic understanding of the interventions across varying pathological stages of the disease. The review also suggests the potential of exploring diverse biomarkers and risk profiles associated with clinically significant outcomes. Moreover, we review findings demonstrating the beneficial effects of PA in various rodent models of cSVD, which have uncovered numerous mechanisms of neuroprotection, including increases in neuroplasticity and integrity of the vasculature and white matter; decreases in inflammation, oxidative stress, and mitochondrial dysfunction; and alterations in amyloid processing and neurotransmitter signaling. In conclusion, this review highlights the potential of physical activity as a preventive strategy for addressing cSVD, offering insights into the need for refining exercise parameters, diversifying research populations, and exploring novel biomarkers, while shedding light on the intricate mechanisms through which exercise confers neuroprotection in both humans and animal models.
Collapse
Affiliation(s)
- Maria E Anderson
- Department of Psychology, Family, and Justice Studies, University of Saint Joseph, 1678 Asylum Ave, West Hartford, CT 06117, USA
| | - Eleanor J Wind
- Department of Psychology and Neuroscience, Nova Southeastern University, 3300 S. University Drive, Fort Lauderdale, FL 33328, USA
| | - Lisa S Robison
- Department of Psychology and Neuroscience, Nova Southeastern University, 3300 S. University Drive, Fort Lauderdale, FL 33328, USA.
| |
Collapse
|
2
|
Bordet S, Luaces JP, Herrera MI, Gonzalez LM, Kobiec T, Perez-Lloret S, Otero-Losada M, Capani F. Neuroprotection from protein misfolding in cerebral hypoperfusion concurrent with metabolic syndrome. A translational perspective. Front Neurosci 2023; 17:1215041. [PMID: 37650104 PMCID: PMC10463751 DOI: 10.3389/fnins.2023.1215041] [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: 05/01/2023] [Accepted: 07/17/2023] [Indexed: 09/01/2023] Open
Abstract
Based on clinical and experimental evidence, metabolic syndrome (MetS) and type 2 diabetes (T2D) are considered risk factors for chronic cerebral hypoperfusion (CCH) and neurodegeneration. Scientific evidence suggests that protein misfolding is a potential mechanism that explains how CCH can lead to either Alzheimer's disease (AD) or vascular cognitive impairment and dementia (VCID). Over the last decade, there has been a significant increase in the number of experimental studies regarding this issue. Using several animal paradigms and different markers of CCH, scientists have discussed the extent to which MetSor T2D causes a decrease in cerebral blood flow (CBF). In addition, different models of CCH have explored how long-term reductions in oxygen and energy supply can trigger AD or VCID via protein misfolding and aggregation. Research that combines two or three animal models could broaden knowledge of the links between these pathological conditions. Recent experimental studies suggest novel neuroprotective properties of protein-remodeling factors. In this review, we present a summarized updated revision of preclinical findings, discussing clinical implications and proposing new experimental approaches from a translational perspective. We are confident that research studies, both clinical and experimental, may find new diagnostic and therapeutic tools to prevent neurodegeneration associated with MetS, diabetes, and any other chronic non-communicable disease (NCD) associated with diet and lifestyle risk factors.
Collapse
Affiliation(s)
- Sofía Bordet
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas, CAECIHS, UAI-CONICET, Buenos Aires, Argentina
- Centro de Investigaciones en Psicología y Psicopedagogía (CIPP), Facultad de Psicología y Psicopedagogía, Pontificia Universidad Católica Argentina (UCA), Buenos Aires, Argentina
| | - Juan Pablo Luaces
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas, CAECIHS, UAI-CONICET, Buenos Aires, Argentina
| | - Maria Ines Herrera
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas, CAECIHS, UAI-CONICET, Buenos Aires, Argentina
- Centro de Investigaciones en Psicología y Psicopedagogía (CIPP), Facultad de Psicología y Psicopedagogía, Pontificia Universidad Católica Argentina (UCA), Buenos Aires, Argentina
| | - Liliana Mirta Gonzalez
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas, CAECIHS, UAI-CONICET, Buenos Aires, Argentina
| | - Tamara Kobiec
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas, CAECIHS, UAI-CONICET, Buenos Aires, Argentina
- Centro de Investigaciones en Psicología y Psicopedagogía (CIPP), Facultad de Psicología y Psicopedagogía, Pontificia Universidad Católica Argentina (UCA), Buenos Aires, Argentina
| | - Santiago Perez-Lloret
- Departamento de Fisiología, Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Observatorio de Salud Pública, Pontificia Universidad Católica Argentina, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Matilde Otero-Losada
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas, CAECIHS, UAI-CONICET, Buenos Aires, Argentina
| | - Francisco Capani
- Centro de Altos Estudios en Ciencias Humanas y de la Salud, Universidad Abierta Interamericana, Consejo Nacional de Investigaciones Científicas y Técnicas, CAECIHS, UAI-CONICET, Buenos Aires, Argentina
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago, Chile
| |
Collapse
|
3
|
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
|
4
|
Leyder E, Suresh P, Jun R, Overbey K, Banerjee T, Melnikova T, Savonenko A. Depression-related phenotypes at early stages of Aβ and tau accumulation in inducible Alzheimer's disease mouse model: Task-oriented and concept-driven interpretations. Behav Brain Res 2023; 438:114187. [PMID: 36343696 DOI: 10.1016/j.bbr.2022.114187] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 10/16/2022] [Accepted: 10/30/2022] [Indexed: 11/06/2022]
Abstract
Depression is highly prevalent in Alzheimer Disease (AD); however, there is paucity of studies that focus specifically on the assessment of depression-relevant phenotypes in AD mouse models. Conditional doxycycline-dependent transgenic mouse models reproducing amyloidosis (TetOffAPPsi) and/or tau (TetOffTauP301L) pathology starting at middle age (6 months) were used in this study. As AD patients can experience depressive symptoms relatively early in disease, testing was conducted at early, pre-pathology stages of Aβ and/or tau accumulation (starting from 45 days of transgenes expression). Tau-related differences were detected in the Novelty Suppressed Feeding task (NSF), whereas APP-related differences were observed predominantly in measures of the Open Field (OF) and Forced Swim tasks (FST). Effects of combined production of Aβ and tau were detected in immobility during the 1st half of the Tail Suspension task (TST). These data demonstrate that results from different tasks are difficult to reconcile using task/variable-centered interpretations in which a single task/variable is assigned an ad-hoc meaning relevant to depression. An alternative, concept-oriented, approach is based on multiple variables/tests, with an understanding of their possible inter-dependence and utilization of statistical approaches that handle correlated data sets. The existence of strong correlations within and between some of the tasks supported utilization of factor analyses (FA). FA explained a similar amount of variability across the genotypes (∼80%) and identified two factors stable across genotypes and representing motor activity and anxiety measures in OF. In contrast, variables related to FST, TST, and NSFT did not demonstrate a structure of factor loadings that would support the existence of a single integral factor of "depressive state" measured by these tasks. In addition, factor loadings varied between genotypes, indicating that genotype-specific between-task correlations need to be considered for interpretations of findings in any single task. In general, this study demonstrates that utilization of multiple tasks to characterize behavioral phenotypes, an approach that is finally gaining more widespread adoption, requires a step of data integration across different behavioral tests for appropriate interpretations.
Collapse
Affiliation(s)
- Erica Leyder
- Department of Pathology, The Johns Hopkins University School of Medicine, 558 Ross Research Building, 720 Rutland Avenue, Baltimore, MD 21205, USA
| | - Prakul Suresh
- Department of Pathology, The Johns Hopkins University School of Medicine, 558 Ross Research Building, 720 Rutland Avenue, Baltimore, MD 21205, USA
| | - Rachel Jun
- Department of Pathology, The Johns Hopkins University School of Medicine, 558 Ross Research Building, 720 Rutland Avenue, Baltimore, MD 21205, USA
| | - Katherine Overbey
- Department of Pathology, The Johns Hopkins University School of Medicine, 558 Ross Research Building, 720 Rutland Avenue, Baltimore, MD 21205, USA
| | - Tirtho Banerjee
- Department of Pathology, The Johns Hopkins University School of Medicine, 558 Ross Research Building, 720 Rutland Avenue, Baltimore, MD 21205, USA
| | - Tatiana Melnikova
- Department of Pathology, The Johns Hopkins University School of Medicine, 558 Ross Research Building, 720 Rutland Avenue, Baltimore, MD 21205, USA.
| | - Alena Savonenko
- Department of Pathology, The Johns Hopkins University School of Medicine, 558 Ross Research Building, 720 Rutland Avenue, Baltimore, MD 21205, USA
| |
Collapse
|
5
|
Biose IJ, Ismael S, Ouvrier B, White AL, Bix GJ. The Potential Role of Integrin Signaling in Memory and Cognitive Impairment. Biomolecules 2023; 13:biom13010108. [PMID: 36671492 PMCID: PMC9855855 DOI: 10.3390/biom13010108] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/29/2022] [Accepted: 01/02/2023] [Indexed: 01/06/2023] Open
Abstract
Dementia currently has no cure and, due to the increased prevalence and associated economic and personal burden of this condition, current research efforts for the development of potential therapies have intensified. Recently, targeting integrins as a strategy to ameliorate dementia and other forms of cognitive impairment has begun to gain traction. Integrins are major bidirectional signaling receptors in mammalian cells, mediating various physiological processes such as cell-cell interaction and cell adhesion, and are also known to bind to the extracellular matrix. In particular, integrins play a critical role in the synaptic transmission of signals, hence their potential contribution to memory formation and significance in cognitive impairment. In this review, we describe the physiological roles that integrins play in the blood-brain barrier (BBB) and in the formation of memories. We also provide a clear overview of how integrins are implicated in BBB disruption following cerebral pathology. Given that vascular contributions to cognitive impairment and dementia and Alzheimer's' disease are prominent forms of dementia that involve BBB disruption, as well as chronic inflammation, we present current approaches shown to improve dementia-like conditions with integrins as a central focus. We conclude that integrins are vital in memory formation and that their disruption could lead to various forms of cognitive impairment. While further research to understand the relationships between integrins and memory is needed, we propose that the translational relevance of research efforts in this area could be improved through the use of appropriately aged, comorbid, male and female animals.
Collapse
Affiliation(s)
- Ifechukwude Joachim Biose
- Clinical Neuroscience Research Center, Department of Neurosurgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Saifudeen Ismael
- Clinical Neuroscience Research Center, Department of Neurosurgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Blake Ouvrier
- Clinical Neuroscience Research Center, Department of Neurosurgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
- Tulane Brain Institute, Tulane University, New Orleans, LA 70112, USA
| | - Amanda Louise White
- Clinical Neuroscience Research Center, Department of Neurosurgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
- Tulane Brain Institute, Tulane University, New Orleans, LA 70112, USA
| | - Gregory Jaye Bix
- Clinical Neuroscience Research Center, Department of Neurosurgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
- School of Medicine, Tulane University, New Orleans, LA 70112, USA
- Department of Neurology, Tulane University School of Medicine, New Orleans, LA 70112, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA
- School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA 70122, USA
- Correspondence: ; Tel.: +1-504-988-3564
| |
Collapse
|
6
|
Chandran R, He L, Nie X, Voltin J, Jamil S, Doueiry C, Falangola MF, Ergul A, Li W. Magnetic resonance imaging reveals microemboli-mediated pathological changes in brain microstructure in diabetic rats: relevance to vascular cognitive impairment/dementia. Clin Sci (Lond) 2022; 136:1555-1570. [PMID: 36314470 PMCID: PMC10066787 DOI: 10.1042/cs20220465] [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: 07/06/2022] [Revised: 10/19/2022] [Accepted: 10/28/2022] [Indexed: 11/17/2022]
Abstract
Diabetes doubles the risk of vascular cognitive impairment, but the underlying reasons remain unclear. In the present study, we determined the temporal and spatial changes in the brain structure after microemboli (ME) injection using diffusion MRI (dMRI). Control and diabetic rats received cholesterol crystal ME (40-70 µm) injections. Cognitive tests were followed up to 16 weeks, while dMRI scans were performed at baseline and 12 weeks post-ME. The novel object recognition test had a lower d2 recognition index along with a decrease in spontaneous alternations in the Y maze test in diabetic rats with ME. dMRI showed that ME injection caused infarction in two diabetic animals (n=5) but none in controls (n=6). In diabetes, radial diffusivity (DR) was increased while fractional anisotropy (FA) was decreased in the cortex, indicating loss of tissue integrity and edema. In the dorsal hippocampus, mean diffusivity (MD), axial diffusivity (DA), and DR were significantly increased, indicating loss of axons and myelin damage. Histological analyses confirmed more tissue damage and microglial activation in diabetic rats with ME. These results suggest that ME injury and associated cerebrovascular dysfunction are greater in diabetes, which may cause cognitive deficits. Strategies to improve vascular function can be a preventive and therapeutic approach for vascular cognitive impairment.
Collapse
Affiliation(s)
- Raghavendar Chandran
- Ralph H. Johnson VA Medical Center, Charleston, SC
- Department of Pathology & Laboratory Medicine, Medical University of South Carolina, Charleston, SC
| | - Lianying He
- Ralph H. Johnson VA Medical Center, Charleston, SC
- Department of Pathology & Laboratory Medicine, Medical University of South Carolina, Charleston, SC
| | - Xingju Nie
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC
- Center for Biomedical Imaging, Medical University of South Carolina, Charleston, SC
| | - Joshua Voltin
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC
- Center for Biomedical Imaging, Medical University of South Carolina, Charleston, SC
| | - Sarah Jamil
- Ralph H. Johnson VA Medical Center, Charleston, SC
- Department of Pathology & Laboratory Medicine, Medical University of South Carolina, Charleston, SC
| | - Caren Doueiry
- Ralph H. Johnson VA Medical Center, Charleston, SC
- Department of Pathology & Laboratory Medicine, Medical University of South Carolina, Charleston, SC
| | - Maria Fatima Falangola
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC
- Center for Biomedical Imaging, Medical University of South Carolina, Charleston, SC
| | - Adviye Ergul
- Ralph H. Johnson VA Medical Center, Charleston, SC
- Department of Pathology & Laboratory Medicine, Medical University of South Carolina, Charleston, SC
| | - Weiguo Li
- Ralph H. Johnson VA Medical Center, Charleston, SC
- Department of Pathology & Laboratory Medicine, Medical University of South Carolina, Charleston, SC
| |
Collapse
|
7
|
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: 23] [Impact Index Per Article: 7.7] [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
|
8
|
Zheng L, Jia J, Chen Y, Liu R, Cao R, Duan M, Zhang M, Xu Y. Pentoxifylline alleviates ischemic white matter injury through up-regulating Mertk-mediated myelin clearance. J Neuroinflammation 2022; 19:128. [PMID: 35642056 PMCID: PMC9153105 DOI: 10.1186/s12974-022-02480-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 05/15/2022] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Vascular dementia (VAD) is the second most common type of dementia lacking effective treatments. Pentoxifylline (PTX), a nonselective phosphodiesterase inhibitor, displays protective effects in multiple cerebral diseases. In this study, we aimed to investigate the therapeutic effects and potential mechanisms of PTX in VAD. METHODS Bilateral common carotid artery stenosis (BCAS) mouse model was established to mimic VAD. Mouse behavior was tested by open field test, novel object recognition test, Y-maze and Morris water maze (MWM) tests. Histological staining, magnetic resonance imaging (MRI) and electron microscopy were used to define white matter integrity. The impact of PTX on microglia phagocytosis, peroxisome proliferator-activated receptors-γ (PPAR-γ) activation and Mer receptor tyrosine kinase (Mertk) expression was assessed by immunofluorescence, western blotting and flow cytometry with the application of microglia-specific Mertk knockout mice, Mertk inhibitor and PPAR-γ inhibitor. RESULTS Here, we found that PTX treatment alleviated cognitive impairment in novel object recognition test, Y-maze and Morris water maze tests. Furthermore, PTX alleviated white matter injury in corpus callosum (CC) and internal capsule (IC) areas as shown by histological staining and MRI analysis. PTX-treatment group presented thicker myelin sheath than vehicle group by electron microscopy. Mechanistically, PTX facilitated microglial phagocytosis of myelin debris by up-regulating the expression of Mertk in BCAS model and primary cultured microglia. Importantly, microglia-specific Mertk knockout blocked the therapeutic effects of PTX in BCAS model. Moreover, Mertk expression was regulated by the nuclear translocation of PPAR-γ. Through modulating PPAR-γ, PTX enhanced Mertk expression. CONCLUSIONS Collectively, our results demonstrated that PTX showed therapeutic potentials in VAD and alleviated ischemic white matter injury via modulating Mertk-mediated myelin clearance in microglia.
Collapse
Affiliation(s)
- Lili Zheng
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing University Medical School, 321 ZhongShan Road, Nanjing, 210008, Jiangsu, China
| | - Junqiu Jia
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing University Medical School, 321 ZhongShan Road, Nanjing, 210008, Jiangsu, China
| | - Yan Chen
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing University Medical School, 321 ZhongShan Road, Nanjing, 210008, Jiangsu, China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China
- Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, China
- Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, China
| | - Renyuan Liu
- Department of Radiology, The Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Runjing Cao
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing University Medical School, 321 ZhongShan Road, Nanjing, 210008, Jiangsu, China
| | - Manlin Duan
- Department of Anesthesiology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China.
| | - Meijuan Zhang
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing University Medical School, 321 ZhongShan Road, Nanjing, 210008, Jiangsu, China.
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China.
- Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, China.
- Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, China.
| | - Yun Xu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing University Medical School, 321 ZhongShan Road, Nanjing, 210008, Jiangsu, China.
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China.
- Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, China.
- Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, China.
| |
Collapse
|
9
|
Wei B, Wu S, Wang Z, Song W, Zhu J. Comparison of Cognitive Performance and Cardiac Function Between Three Different Rat Models of Vascular Dementia. Neuropsychiatr Dis Treat 2022; 18:19-28. [PMID: 35018098 PMCID: PMC8742620 DOI: 10.2147/ndt.s338226] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 11/08/2021] [Indexed: 01/01/2023] Open
Abstract
PURPOSE Establishing an ideal animal model is essential for studying the pathogenesis, prevention and treatment of vascular dementia (VD). The present study was designed to compare the differences of behavior, cerebral blood flow (CBF), cardiac output and the levels of myocardial enzyme of three different VD rat models. METHODS The rats were randomly divided into sham-operated group (SHAM), permanent bilateral common carotid artery occlusion group (BCCAO), BCCAO combined with sodium nitroprusside (2.0mg·kg-1) group (BCCAO+2.0SNP) and BCCAO combined with sodium nitroprusside (2.5mg·kg-1) group (BCCAO+2.5SNP). After operation, Morris water maze test, echocardiographic evaluation and the measurement of CBF were performed, then the levels of myocardial enzymes in serum were assessed during euthanasia. RESULTS Compared with SHAM rats, the three VD model rats showed different degrees of cognitive impairment, lower cardiac output and CBF, and BCCAO rats showed higher levels of myocardial enzymes. Compared with BCCAO rats, the spatial learning ability of BCCAO+2.0SNP rats and BCCAO+2.5SNP rats was more severely impaired, while the levels of myocardial enzymes of BCCAO+2.0SNP rats were lower. Compared with BCCAO+2.0SNP rats, BCCAO+2.5SNP rats showed no significant difference in cognitive function and cardiac function. CONCLUSION Our present study demonstrated that all of the three different VD rat models exhibited cognitive and cardiac function impairment. The BCCAO+2.0SNP model and BCCAO+2.5SNP model damaged the spatial learning ability more seriously. The BCCAO+2.5SNP model caused more comprehensive cognitive impairment. In addition, the BCCAO+2.0SNP model and BCCAO+2.5SNP model might cause more serious damage to cardiac function.
Collapse
Affiliation(s)
- Baoyu Wei
- State Key Laboratory of Component-Based Chinese Medicine, Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China
| | - Shihao Wu
- State Key Laboratory of Component-Based Chinese Medicine, Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China
| | - Zhaoqi Wang
- State Key Laboratory of Component-Based Chinese Medicine, Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China
| | - Wanshan Song
- Department of Acupuncture and Cerebropathy, Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300250, People's Republic of China
| | - Jinqiang Zhu
- State Key Laboratory of Component-Based Chinese Medicine, Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China
| |
Collapse
|
10
|
Washida K, Kitajima E, Tanaka T, Ikeda S, Chiba T, Noda K, Yoshimoto T, Fukuma K, Saito S, Ihara M. A Nationwide Multi-Center Questionnaire Survey on the Real-World State and Clinical Management of Poststroke Dementia in Japan. J Alzheimers Dis 2021; 84:1103-1114. [PMID: 34633324 PMCID: PMC8673533 DOI: 10.3233/jad-215006] [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] [Indexed: 11/20/2022]
Abstract
Background: Poststroke dementia (PSD) is a serious problem for stroke survivors. However, there is still limited data on the real-world state and clinical management of PSD worldwide, and several countries already have a super-aged society. Objective: We conducted a nationwide questionnaire survey to examine the real-world state and management of PSD in Japan. Methods: A survey was conducted in the top 500 Japanese hospitals regarding the number of stroke patients treated between July 2018 and August 2019. Thirteen questions regarding PSD were mailed to doctors responsible for stroke management. Results: Responses were obtained from 251 hospitals (50.2%). The chief doctors responsible for stroke management answered the questionnaires. The median numbers of patients admitted annually with stroke in the departments of neurology and neurosurgery in the hospitals were 281.0 (interquartile range [IQR], 231.8–385.3) and 253.5 (IQR, 210.0–335.3), respectively, and most hospitals were acute care hospitals. Executive dysfunction was the most common cognitive dysfunction (10.9%), followed by amnesia (9.5%) and apathy (4.1%). Surprisingly, many stroke survivors lived alone at home (23.7%). Montreal Cognitive Assessment was significantly uncommon compared to Mini-Mental State Examination (p < 0.01). Furthermore, objective evaluation tests for behavioral and psychological symptoms of dementia were not often performed. Cognitive rehabilitation treatments were performed more often and earlier than drug treatments. The first drug of choice for PSD was predominantly donepezil (79.1%), followed by galantamine (6.1%), cilostazol (4.9%), memantine (2.5%), and rivastigmine (1.8%). Conclusion: Our study provides real-world evidence for the state of clinical practice related to PSD in Japan.
Collapse
Affiliation(s)
- Kazuo Washida
- Department of Neurology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Erika Kitajima
- Department of Neurology, National Cerebral and Cardiovascular Center, Osaka, Japan.,Department of Medical Engineering, Faculty of Healthcare Sciences, Himeji Dokkyo University, Hyogo, Japan
| | - Tomotaka Tanaka
- Department of Neurology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Shuhei Ikeda
- Department of Neurology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Tetsuya Chiba
- Department of Neurology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Kotaro Noda
- Department of Neurology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Takeshi Yoshimoto
- Department of Neurology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Kazuki Fukuma
- Department of Neurology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Satoshi Saito
- Department of Neurology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Masafumi Ihara
- Department of Neurology, National Cerebral and Cardiovascular Center, Osaka, Japan
| |
Collapse
|
11
|
Li X, Wen W, Li P, Fu Y, Chen H, Wang F, Dai Y, Xu S. Mitochondrial Protection and Against Glutamate Neurotoxicity via Shh/Ptch1 Signaling Pathway to Ameliorate Cognitive Dysfunction by Kaixin San in Multi-Infarct Dementia Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5590745. [PMID: 34306310 PMCID: PMC8285175 DOI: 10.1155/2021/5590745] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 06/17/2021] [Indexed: 12/05/2022]
Abstract
Multi-infarct dementia (MID), a prominent subtype of vascular dementia (VD), is responsible for at least 15 to 20 percent of dementia in the elderly. Mitochondrial dysfunctions and glutamate neurotoxicity due to chronic hypoperfusion and oxidative stress were regarded as the major risk factors in the pathogenesis. Kaixin San (KXS), a classic prescription of Beiji Qianjin Yaofang, was applied to treatment for "amnesia" and has been demonstrated to alleviate the cognitive deficit in a variety of dementias, including MID. However, little is known whether mitochondria and glutamate are associated with the protection of KXS in MID treatment. The aim of this study was to investigate the role of KXS in improving the cognitive function of MID rats through strengthening mitochondrial functions and antagonizing glutamate neurotoxicity via the Shh/Ptch1 signaling pathway. Our data showed that KXS significantly ameliorated memory impairment and hippocampal neuron damage in MID rats. Moreover, KXS improved hippocampal mitochondrial functions by reducing the degree of mitochondrial swelling, increasing the mitochondrial membrane potential (MMP), and elevating the energy charge (EC) and ATP content in MID rats. As expected, the concentration of glutamate and the expression of p-NMDAR1 were significantly reduced by KXS in the brain tissue of MID rats. Furthermore, our results showed that KXS noticeably activated the Shh/Ptch1 signaling pathway which was demonstrated by remarkable elevations of Ptch1, Smo, and Gli1 protein levels in the brain tissue of MID rats. Intriguingly, the inhibition of the Shh signaling pathway with cyclopamine significantly inhibited the protective effects of KXS on glutamate-induced neurotoxicity in PC12 cells. To sum up, these findings suggested that KXS protected MID rats from memory loss by rescuing mitochondrial functions as well as against glutamate neurotoxicity through activating Shh/Ptch1 signaling pathway.
Collapse
Affiliation(s)
- Xiaoqiong Li
- Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
| | - Wen Wen
- Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
| | - Ping Li
- Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
| | - Ying Fu
- Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
| | - Hao Chen
- Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
| | - Fushun Wang
- Institute of Brain and Psychological Science, Sichuan Normal University, Chengdu 610060, China
| | - Yuan Dai
- Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, China
| | - Shijun Xu
- Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu, Sichuan 611137, China
| |
Collapse
|
12
|
Duong MT, Nasrallah IM, Wolk DA, Chang CCY, Chang TY. Cholesterol, Atherosclerosis, and APOE in Vascular Contributions to Cognitive Impairment and Dementia (VCID): Potential Mechanisms and Therapy. Front Aging Neurosci 2021; 13:647990. [PMID: 33841127 PMCID: PMC8026881 DOI: 10.3389/fnagi.2021.647990] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 03/08/2021] [Indexed: 12/18/2022] Open
Abstract
Vascular contributions to cognitive impairment and dementia (VCID) are a common cause of cognitive decline, yet limited therapies exist. This cerebrovascular disease results in neurodegeneration via acute, chronic, local, and systemic mechanisms. The etiology of VCID is complex, with a significant impact from atherosclerosis. Risk factors including hypercholesterolemia and hypertension promote intracranial atherosclerotic disease and carotid artery stenosis (CAS), which disrupt cerebral blood flow and trigger ischemic strokes and VCID. Apolipoprotein E (APOE) is a cholesterol and phospholipid carrier present in plasma and various tissues. APOE is implicated in dyslipidemia and Alzheimer disease (AD); however, its connection with VCID is less understood. Few experimental models for VCID exist, so much of the present information has been drawn from clinical studies. Here, we review the literature with a focus on the clinical aspects of atherosclerotic cerebrovascular disease and build a working model for the pathogenesis of VCID. We describe potential intermediate steps in this model, linking cholesterol, atherosclerosis, and APOE with VCID. APOE4 is a minor isoform of APOE that promotes lipid dyshomeostasis in astrocytes and microglia, leading to chronic neuroinflammation. APOE4 disturbs lipid homeostasis in macrophages and smooth muscle cells, thus exacerbating systemic inflammation and promoting atherosclerotic plaque formation. Additionally, APOE4 may contribute to stromal activation of endothelial cells and pericytes that disturb the blood-brain barrier (BBB). These and other risk factors together lead to chronic inflammation, atherosclerosis, VCID, and neurodegeneration. Finally, we discuss potential cholesterol metabolism based approaches for future VCID treatment.
Collapse
Affiliation(s)
- Michael Tran Duong
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Ilya M Nasrallah
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - David A Wolk
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | | | - Ta-Yuan Chang
- Geisel School of Medicine, Dartmouth College, Hanover, NH, United States
| |
Collapse
|
13
|
Liu Y, Cai J, Qu L. Carotid Occlusion Accentuates Aortic Stenosis and Cardiac Remodeling With Preserved Systolic Function in LDL Receptor-Deficient Mice. Front Physiol 2021; 11:578722. [PMID: 33584325 PMCID: PMC7873957 DOI: 10.3389/fphys.2020.578722] [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: 07/01/2020] [Accepted: 11/25/2020] [Indexed: 11/13/2022] Open
Abstract
Background: Carotid atherosclerotic disease is associated with aortic stenosis and reduced cardiac function. The causality between carotid and cardiac pathologies is unknown. We aim to explore the effects of carotid stenosis or occlusion on cardiac pathology and function. Methods and Results: We produced carotid obstruction or stenosis in 36 atherogenic mice with 150- or 300-μm tandem surgery or sham surgery. The structure and function of the heart were assessed by histology and animal ultrasound. The 150-μm group had larger plaque burden and thicker valve leaflets in the aortic root than did the control group. Also, the two surgery groups had a thicker left ventricular posterior wall and smaller internal diameter compared with controls. Increased myocardial fibrosis was also found in the 150-μm group compared with controls, although the surgery groups had preserved systolic function compared with that of controls. Conclusions: In a mouse model, carotid occlusion accentuated the formation of aortic stenosis and promoted ventricular remodeling without impairing systolic function. Carotid atherosclerotic plaque may be a pathogenic factor for aortic stenosis and ventricular remodeling.
Collapse
Affiliation(s)
- Yandong Liu
- Department of Vascular and Endovascular Surgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Jiawei Cai
- Department of Vascular and Endovascular Surgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Lefeng Qu
- Department of Vascular and Endovascular Surgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
14
|
Cui Y, Jin X, Choi JY, Kim BG. Modeling subcortical ischemic white matter injury in rodents: unmet need for a breakthrough in translational research. Neural Regen Res 2021; 16:638-642. [PMID: 33063714 PMCID: PMC8067929 DOI: 10.4103/1673-5374.295313] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Subcortical ischemic white matter injury (SIWMI), pathological correlate of white matter hyperintensities or leukoaraiosis on magnetic resonance imaging, is a common cause of cognitive decline in elderly. Despite its high prevalence, it remains unknown how various components of the white matter degenerate in response to chronic ischemia.This incomplete knowledge is in part due to a lack of adequate animal model. The current review introduces various SIWMI animal models and aims to scrutinize their advantages and disadvantages primarily in regard to the pathological manifestations of white matter components. The SIWMI animal models are categorized into 1) chemically induced SIWMI models, 2) vascular occlusive SIWMI models, and 3) SIWMI models with comorbid vascular risk factors. Chemically induced models display consistent lesions in predetermined areas of the white matter, but the abrupt evolution of lesions does not appropriately reflect the progressive pathological processes in human white matter hyperintensities. Vascular occlusive SIWMI models often do not exhibit white matter lesions that are sufficiently unequivocal to be quantified. When combined with comorbid vascular risk factors (specifically hypertension), however, they can produce progressive and definitive white matter lesions including diffuse rarefaction, demyelination, loss of oligodendrocytes, and glial activation, which are by far the closest to those found in human white matter hyperintensities lesions. However, considerable surgical mortality and unpredictable natural deaths during a follow-up period would necessitate further refinements in these models. In the meantime, in vitro SIWMI models that recapitulate myelinated white matter track may be utilized to study molecular mechanisms of the ischemic white matter injury. Appropriate in vivo and in vitro SIWMI models will contribute in a complementary manner to making a breakthrough in developing effective treatment to prevent progression of white matter hyperintensities.
Collapse
Affiliation(s)
- Yuexian Cui
- Department of Brain Science, Ajou University School of Medicine; Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Korea; Department of Neurology, Yanbian University Hospital, Yanji, Jilin Province, China
| | - Xuelian Jin
- Department of Brain Science, Ajou University School of Medicine; Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Korea; Department of Nephrology, Suqian First Hospital, Suqian, Jiangsu Province, China
| | - Jun Young Choi
- Department of Brain Science, Ajou University School of Medicine; Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University Graduate School of Medicine; Department of Neurology, Ajou University School of Medicine, Suwon, Korea
| | - Byung Gon Kim
- Department of Brain Science, Ajou University School of Medicine; Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University Graduate School of Medicine; Department of Neurology, Ajou University School of Medicine, Suwon, Korea
| |
Collapse
|
15
|
Brain Functional Network in Chronic Asymptomatic Carotid Artery Stenosis and Occlusion: Changes and Compensation. Neural Plast 2020; 2020:9345602. [PMID: 33029129 PMCID: PMC7530486 DOI: 10.1155/2020/9345602] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 09/09/2020] [Indexed: 11/17/2022] Open
Abstract
Asymptomatic carotid artery stenosis (CAS) and occlusion (CAO) disrupt cerebral hemodynamics. There are few studies on the brain network changes and compensation associated with the progression from chronic CAS to CAO. In the current study, our goal is to improve the understanding of the specific abnormalities and compensatory phenomena associated with the functional connection in patients with CAS and CAO. In this prospective study, 27 patients with CAO, 29 patients with CAS, and 15 healthy controls matched for age, sex, education, handedness, and risk factors underwent neuropsychological testing and resting-state functional magnetic resonance (rs-fMRI) imaging simultaneously; graph theoretical analysis of brain networks was performed to determine the relationship between changes in brain network connectivity and the progression from internal CAS to CAO. The global properties of the brain network assortativity (p = 0.002), hierarchy (p = 0.002), network efficiency (p = 0.011), and small-worldness (p = 0.009) were significantly more abnormal in the CAS group than in the control and CAO groups. In patients with CAS and CAO, the nodal efficiency of key nodes in multiple brain regions decreased, while the affected hemisphere lost many key functional connections. In this study, we found that patients with CAS showed grade reconstruction, invalid connections, and other phenomena that impaired the efficiency of information transmission in the brain network. A compensatory functional connection in the contralateral cerebral hemisphere of patients with CAS and CAO may be an important mechanism that maintains clinical asymptomatic performance. This study not only reveals the compensation mechanism of cerebral hemisphere ischemia but also validates previous explanations for brain function connectivity, which can help provide interventions in advance and reduce the impairment of higher brain functions. This trial is registered with Clinical Trial Registration-URL http://www.chictr.org.cn and Unique identifier ChiCTR1900023610.
Collapse
|
16
|
Bhatia P, Singh N. Tadalafil ameliorates memory deficits, oxidative stress, endothelial dysfunction and neuropathological changes in rat model of hyperhomocysteinemia induced vascular dementia. Int J Neurosci 2020; 132:384-396. [PMID: 32859137 DOI: 10.1080/00207454.2020.1817009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
AIM The present study investigates the potential of Tadalafil, a phosphodiesterase-5 inhibitor, in a rat model of hyperhomocysteinemia induced vascular dementia. METHODS Hyperhomocysteinemia induced vascular dementia in Wistar rats was produced by administering l-Methionine (1.7 g/kg/day; p.o.×8 weeks). Learning and memory was assessed by employing Morris water maze (MWM) test. Endothelial dysfunction was assessed through acetylcholine-induced endothelial-dependent vasorelaxation and serum nitrite levels. Various other biochemical and histopathological estimations were also performed. RESULTS l-Methionine produced significant impairment in acetylcholine-induced endothelium-dependent vasorelaxation and a decrease in serum nitrite levels indicating endothelial dysfunction. Further, these animals performed poorly on Morris water maze, depicting impairment of learning and memory. There was a significant rise in brain oxidative stress level (indicated by an increase in brain thiobarbituric acid reactive species and a decrease in reduced glutathione levels). Increase in brain acetylcholinesterase activity; brain myeloperoxidase activity and brain neutrophil infiltration (a marker of inflammation) were also observed. Tadalafil (5 and 10 mg/kg, p.o.)/Donepezil (0.5 mg/kg, i.p., serving as standard) treatment ameliorated l-Methionine induced endothelial dysfunction; memory deficits; biochemical and histopathological changes in a significant manner. CONCLUSIONS It may be concluded that tadalafil has shown efficacy in the rat model of l-Methionine induced vascular dementia and that phosphodiesterase-5 can be considered as an important therapeutic target for the treatment of vascular dementia.
Collapse
Affiliation(s)
- Pankaj Bhatia
- CNS Research Lab., Pharmacology Division, Department of Pharmaceutical Sciences and Drug Research, Faculty of Medicine, Punjabi University, Patiala, Punjab, India
| | - Nirmal Singh
- Department of Pharmaceutical Sciences and Drug Research, Faculty of Medicine, Punjabi University, Patiala, Punjab, India
| |
Collapse
|
17
|
Carare RO, Aldea R, Agarwal N, Bacskai BJ, Bechman I, Boche D, Bu G, Bulters D, Clemens A, Counts SE, de Leon M, Eide PK, Fossati S, Greenberg SM, Hamel E, Hawkes CA, Koronyo‐Hamaoui M, Hainsworth AH, Holtzman D, Ihara M, Jefferson A, Kalaria RN, Kipps CM, Kanninen KM, Leinonen V, McLaurin J, Miners S, Malm T, Nicoll JAR, Piazza F, Paul G, Rich SM, Saito S, Shih A, Scholtzova H, Snyder H, Snyder P, Thormodsson FR, van Veluw SJ, Weller RO, Werring DJ, Wilcock D, Wilson MR, Zlokovic BV, Verma A. Clearance of interstitial fluid (ISF) and CSF (CLIC) group-part of Vascular Professional Interest Area (PIA): Cerebrovascular disease and the failure of elimination of Amyloid-β from the brain and retina with age and Alzheimer's disease-Opportunities for Therapy. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2020; 12:e12053. [PMID: 32775596 PMCID: PMC7396859 DOI: 10.1002/dad2.12053] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 05/26/2020] [Indexed: 12/22/2022]
Abstract
Two of the key functions of arteries in the brain are (1) the well-recognized supply of blood via the vascular lumen and (2) the emerging role for the arterial walls as routes for the elimination of interstitial fluid (ISF) and soluble metabolites, such as amyloid beta (Aβ), from the brain and retina. As the brain and retina possess no conventional lymphatic vessels, fluid drainage toward peripheral lymph nodes is mediated via transport along basement membranes in the walls of capillaries and arteries that form the intramural peri-arterial drainage (IPAD) system. IPAD tends to fail as arteries age but the mechanisms underlying the failure are unclear. In some people this is reflected in the accumulation of Aβ plaques in the brain in Alzheimer's disease (AD) and deposition of Aβ within artery walls as cerebral amyloid angiopathy (CAA). Knowledge of the dynamics of IPAD and why it fails with age is essential for establishing diagnostic tests for the early stages of the disease and for devising therapies that promote the clearance of Aβ in the prevention and treatment of AD and CAA. This editorial is intended to introduce the rationale that has led to the establishment of the Clearance of Interstitial Fluid (ISF) and CSF (CLIC) group, within the Vascular Professional Interest Area of the Alzheimer's Association International Society to Advance Alzheimer's Research and Treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Diederik Bulters
- University of SouthamptonSouthamptonUK
- University Hospital Southampton NHS TrustSouthamptonUK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Christopher M. Kipps
- University of SouthamptonSouthamptonUK
- University Hospital Southampton NHS TrustSouthamptonUK
| | | | | | | | | | - Tarja Malm
- University of Eastern FinlandKuopioFinland
| | | | | | | | | | - Satoshi Saito
- National Cerebral and Cardiovascular CenterOsakaJapan
| | - Andy Shih
- Seattle Children's HospitalSeattleWashingtonUSA
| | | | | | - Peter Snyder
- University of Rhode IslandSouth KingstownRhode IslandUSA
| | | | | | | | - David J. Werring
- Stroke Research CentreUCL Queen Square Institute of NeurologyUniversity College LondonLondonUK
| | | | | | | | - Ajay Verma
- CODIAK BiosciencesCambridgeMassachusettsUSA
| |
Collapse
|
18
|
Lee NK, Kim H, Chang JW, Jang H, Kim H, Yang J, Kim J, Son JP, Na DL. Exploring the Potential of Mesenchymal Stem Cell-Based Therapy in Mouse Models of Vascular Cognitive Impairment. Int J Mol Sci 2020; 21:ijms21155524. [PMID: 32752272 PMCID: PMC7432487 DOI: 10.3390/ijms21155524] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/25/2020] [Accepted: 07/30/2020] [Indexed: 12/29/2022] Open
Abstract
Closely linked to Alzheimer’s disease (AD), the pathological spectrum of vascular cognitive impairment (VCI) is known to be wide and complex. Considering that multiple instead of a single targeting approach is considered a treatment option for such complicated diseases, the multifaceted aspects of mesenchymal stem cells (MSCs) make them a suitable candidate to tackle the heterogeneity of VCI. MSCs were delivered via the intracerebroventricular (ICV) route in mice that were subjected to VCI by carotid artery stenosis. VCI was induced in C57BL6/J mice wild type (C57VCI) mice by applying a combination of ameroid constrictors and microcoils, while ameroid constrictors alone were bilaterally applied to 5xFAD (transgenic AD mouse model) mice (5xVCI). Compared to the controls (minimal essential medium (MEM)-injected C57VCI mice), changes in spatial working memory were not noted in the MSC-injected C57VCI mice, and unexpectedly, the mortality rate was higher. In contrast, compared to the MEM-injected 5xVCI mice, mortality was not observed, and the spatial working memory was also improved in MSC-injected 5xVCI mice. Disease progression of the VCI-induced mice seems to be affected by the method of carotid artery stenosis and due to this heterogeneity, various factors must be considered to maximize the therapeutic benefits exerted by MSCs. Factors, such as the optimal MSC injection time point, cell concentration, sacrifice time point, and immunogenicity of the transplanted cells, must all be adequately addressed so that MSCs can be appropriately and effectively used as a treatment option for VCI.
Collapse
Affiliation(s)
- Na Kyung Lee
- School of Medicine, Sungkyunkwan University, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Korea;
- Stem Cell & Regenerative Medicine Institute, Samsung Medical Center, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Korea; (H.K.); (J.W.C.); (H.J.)
- Samsung Alzheimer Research Center, Samsung Medical Center, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Korea
| | - Hyeongseop Kim
- Stem Cell & Regenerative Medicine Institute, Samsung Medical Center, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Korea; (H.K.); (J.W.C.); (H.J.)
- Stem Cell Institute, ENCell Co. Ltd., Seoul 06072, Korea
| | - Jong Wook Chang
- Stem Cell & Regenerative Medicine Institute, Samsung Medical Center, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Korea; (H.K.); (J.W.C.); (H.J.)
- Stem Cell Institute, ENCell Co. Ltd., Seoul 06072, Korea
| | - Hyemin Jang
- Stem Cell & Regenerative Medicine Institute, Samsung Medical Center, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Korea; (H.K.); (J.W.C.); (H.J.)
- Samsung Alzheimer Research Center, Samsung Medical Center, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Korea
- Department of Neurology, Sungkyunkwan University School of Medicine, Samsung Medical Center, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Korea
| | - Hunnyun Kim
- Laboratory Animal Research Center, Samsung Biomedical Research Institute, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Korea; (H.K.); (J.Y.); (J.K.)
| | - Jehoon Yang
- Laboratory Animal Research Center, Samsung Biomedical Research Institute, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Korea; (H.K.); (J.Y.); (J.K.)
| | - Jeyun Kim
- Laboratory Animal Research Center, Samsung Biomedical Research Institute, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Korea; (H.K.); (J.Y.); (J.K.)
| | - Jeong Pyo Son
- Laboratory Animal Center, Osong Medical Innovation Foundation, Cheongju 28160, Korea;
| | - Duk L. Na
- School of Medicine, Sungkyunkwan University, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Korea;
- Stem Cell & Regenerative Medicine Institute, Samsung Medical Center, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Korea; (H.K.); (J.W.C.); (H.J.)
- Samsung Alzheimer Research Center, Samsung Medical Center, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Korea
- Department of Neurology, Sungkyunkwan University School of Medicine, Samsung Medical Center, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Korea
- Neuroscience Center, Samsung Medical Center, 81 Irwon-ro, Gangnam-gu, Seoul 06351, Korea
- Correspondence: ; Tel.: +82-2-3410-3591; Fax: +82-2-3412-3423
| |
Collapse
|
19
|
Kandasamy M, Anusuyadevi M, Aigner KM, Unger MS, Kniewallner KM, de Sousa DMB, Altendorfer B, Mrowetz H, Bogdahn U, Aigner L. TGF-β Signaling: A Therapeutic Target to Reinstate Regenerative Plasticity in Vascular Dementia? Aging Dis 2020; 11:828-850. [PMID: 32765949 PMCID: PMC7390515 DOI: 10.14336/ad.2020.0222] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 02/22/2020] [Indexed: 12/11/2022] Open
Abstract
Vascular dementia (VaD) is the second leading form of memory loss after Alzheimer's disease (AD). Currently, there is no cure available. The etiology, pathophysiology and clinical manifestations of VaD are extremely heterogeneous, but the impaired cerebral blood flow (CBF) represents a common denominator of VaD. The latter might be the result of atherosclerosis, amyloid angiopathy, microbleeding and micro-strokes, together causing blood-brain barrier (BBB) dysfunction and vessel leakage, collectively originating from the consequence of hypertension, one of the main risk factors for VaD. At the histopathological level, VaD displays abnormal vascular remodeling, endothelial cell death, string vessel formation, pericyte responses, fibrosis, astrogliosis, sclerosis, microglia activation, neuroinflammation, demyelination, white matter lesions, deprivation of synapses and neuronal loss. The transforming growth factor (TGF) β has been identified as one of the key molecular factors involved in the aforementioned various pathological aspects. Thus, targeting TGF-β signaling in the brain might be a promising therapeutic strategy to mitigate vascular pathology and improve cognitive functions in patients with VaD. This review revisits the recent understanding of the role of TGF-β in VaD and associated pathological hallmarks. It further explores the potential to modulate certain aspects of VaD pathology by targeting TGF-β signaling.
Collapse
Affiliation(s)
- Mahesh Kandasamy
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India.
- Faculty Recharge Programme, University Grants Commission (UGC-FRP), New Delhi, India.
| | - Muthuswamy Anusuyadevi
- Molecular Gerontology Group, Department of Biochemistry, School of Life Sciences, Bharathidhasan University, Tiruchirappalli, Tamil Nadu, India.
| | - Kiera M Aigner
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
| | - Michael S Unger
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
| | - Kathrin M Kniewallner
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
| | - Diana M Bessa de Sousa
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
| | - Barbara Altendorfer
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
| | - Heike Mrowetz
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
| | - Ulrich Bogdahn
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
- Velvio GmbH, Regensburg, Germany.
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine, Salzburg, Paracelsus Medical University.
- Spinal Cord Injury and Tissue Regeneration Center, Salzburg, Paracelsus Medical University, Salzburg, Austria.
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
20
|
Niu HM, Ma DL, Wang MY, Chen XP, Zhang L, Li YL, Zhang L, Li L. Epimedium flavonoids protect neurons and synapses in the brain via activating NRG1/ErbB4 and BDNF/Fyn signaling pathways in a chronic cerebral hypoperfusion rat model. Brain Res Bull 2020; 162:132-140. [PMID: 32592805 DOI: 10.1016/j.brainresbull.2020.06.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 06/02/2020] [Accepted: 06/19/2020] [Indexed: 12/20/2022]
Abstract
Cerebral hypoperfusion is a common feature of cerebral small vascular disease (CSVD), which has been considered as one of the causes of cognitive decline in recent years. Epimedium flavonoids (EF) are the main ingredients extracted from Epimedium. The purpose of this study was to investigate the effects of EF on cognitive impairment, and the underlying mechanisms in rats with permanent occlusion of the bilateral common carotid artery (2VO). EF (50, 100, and 200 mg/kg) was intragastrically administered for 12 weeks starting 2 weeks after 2VO surgery. The results showed that EF treatment improved learning and memory impairment in 2VO rats evaluated by novel object recognition and Y-maze tests. NeuN immunohistochemical staining indicated that EF alleviated neuronal loss in the hippocampus and cerebral cortex of 2VO rats. MAP-2 immunofluorescence staining and western blotting showed that EF protected neuronal dendrites and increased the expression of cytoskeleton proteins MAP-2 and NF200 in the hippocampus of 2VO rats. Moreover, EF protected the synapse ultrastructure detected by transmission electron microscopy, and increased the expression of synaptic plasticity-related proteins, including synaptophysin, synaptotagmin-I, synapsin I, PSD-95, p-NMDA2B, and p-CaMKII-α in the hippocampus of 2VO rats. In addition, EF increased the expression of neuregulin-1 (NRG-1), p-ErbB4, brain-derived neurotrophic factor (BDNF), p-Fyn, PI3K, p-Akt, and p-CREB in the hippocampus of 2VO rats. These results suggest that EF may protect neurons and synapses by activating the NRG1/ErbB4, BDNF/Fyn, and P13 K/Akt/CREB pathways in the hippocampus and cerebral cortex, thus improving cognitive impairment induced by chronic cerebral hypoperfusion. EF may be a potential candidate drug for chronic cerebral hypoperfusion and CSVD therapy.
Collapse
Affiliation(s)
- Hong-Mei Niu
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nerve System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, 100053, China
| | - Deng-Lei Ma
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nerve System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, 100053, China
| | - Ming-Yang Wang
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nerve System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, 100053, China
| | - Xiao-Ping Chen
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nerve System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, 100053, China
| | - Li Zhang
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nerve System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, 100053, China
| | - Ya-Li Li
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nerve System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, 100053, China
| | - Lan Zhang
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nerve System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, 100053, China
| | - Lin Li
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nerve System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, 100053, China.
| |
Collapse
|
21
|
Heterogeneous Disease Progression in a Mouse Model of Vascular Cognitive Impairment. Int J Mol Sci 2020; 21:ijms21082820. [PMID: 32316637 PMCID: PMC7215687 DOI: 10.3390/ijms21082820] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/12/2020] [Accepted: 04/15/2020] [Indexed: 11/17/2022] Open
Abstract
Recently, an asymmetric vascular compromise approach that replicates many aspects of human vascular cognitive impairment (VCI) has been reported. The present study aimed to first investigate on the reproducibility in the disease progression of this newly reported VCI model using wild-type C57BL6/J mice. The second aim was to assess how this approach will affect the disease progression of transgenic Alzheimer’s disease (AD) 5XFAD mice subjected to VCI. C57BL6/J and 5XFAD mice were subjected to VCI by placing an ameroid constrictor on the right CCA and a microcoil on the left CCA. Infarcts and hippocampal neuronal loss did not appear predominantly in the right (ameroid side) as expected but randomly in both hemispheres. The mortality rate of C57BL6/J mice was unexpectedly high. Inducing VCI reduced amyloid burden in the hippocampi of 5XFAD mice. Since VCI is known to be complex and complicated, the heterogeneous disease progression observed from this current study shares close resemblance to the clinical manifestation of VCI. This heterogeneity, however, makes it challenging to test novel treatment options using this model. Further study is warranted to tackle the heterogeneous nature of VCI.
Collapse
|
22
|
Role of HMGB1 in an Animal Model of Vascular Cognitive Impairment Induced by Chronic Cerebral Hypoperfusion. Int J Mol Sci 2020; 21:ijms21062176. [PMID: 32245271 PMCID: PMC7139598 DOI: 10.3390/ijms21062176] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 03/20/2020] [Indexed: 01/10/2023] Open
Abstract
The pathophysiology of vascular cognitive impairment (VCI) is associated with chronic cerebral hypoperfusion (CCH). Increased high-mobility group box protein 1 (HMGB1), a nonhistone protein involved in injury and inflammation, has been established in the acute phase of CCH. However, the role of HMGB1 in the chronic phase of CCH remains unclear. We developed a novel animal model of CCH with a modified bilateral common carotid artery occlusion (BCCAO) in C57BL/6 mice. Cerebral blood flow (CBF) reduction, the expression of HMGB1 and its proinflammatory cytokines (tumor necrosis factor-alpha [TNF-α], interleukin [IL]-1β, and IL-6), and brain pathology were assessed. Furthermore, we evaluated the effect of HMGB1 suppression through bilateral intrahippocampus injection with the CRISPR/Cas9 knockout plasmid. Three months after CCH induction, CBF decreased to 30–50% with significant cognitive decline in BCCAO mice. The 7T-aMRI showed hippocampal atrophy, but amyloid positron imaging tomography showed nonsignificant amyloid-beta accumulation. Increased levels of HMGB1, TNF-α, IL-1β, and IL-6 were observed 3 months after BCCAO. HMGB1 suppression with CRISPR/Cas9 knockout plasmid restored TNF-α, IL-1β, and IL-6 and attenuated hippocampal atrophy and cognitive decline. We believe that HMGB1 plays a pivotal role in CCH-induced VCI pathophysiology and can be a potential therapeutic target of VCI.
Collapse
|
23
|
Washida K, Hattori Y, Ihara M. Animal Models of Chronic Cerebral Hypoperfusion: From Mouse to Primate. Int J Mol Sci 2019; 20:ijms20246176. [PMID: 31817864 PMCID: PMC6941004 DOI: 10.3390/ijms20246176] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 11/17/2019] [Accepted: 12/03/2019] [Indexed: 12/15/2022] Open
Abstract
Vascular cognitive impairment (VCI) or vascular dementia occurs as a result of brain ischemia and represents the second most common type of dementia after Alzheimer’s disease. To explore the underlying mechanisms of VCI, several animal models of chronic cerebral hypoperfusion have been developed in rats, mice, and primates. We established a mouse model of chronic cerebral hypoperfusion by narrowing the bilateral common carotid arteries with microcoils, eventually resulting in hippocampal atrophy. In addition, a mouse model of white matter infarct-related damage with cognitive and motor dysfunction has also been established by asymmetric common carotid artery surgery. Although most experiments studying chronic cerebral hypoperfusion have been performed in rodents because of the ease of handling and greater ethical acceptability, non-human primates appear to represent the best model for the study of VCI, due to their similarities in much larger white matter volume and amyloid β depositions like humans. Therefore, we also recently developed a baboon model of VCI through three-vessel occlusion (both the internal carotid arteries and the left vertebral artery). In this review, several animal models of chronic cerebral hypoperfusion, from mouse to primate, are extensively discussed to aid in better understanding of pathophysiology of VCI.
Collapse
Affiliation(s)
- Kazuo Washida
- Correspondence: ; Tel.: +81-6-6170-1070; Fax: +81-6-6170-1782
| | | | | |
Collapse
|
24
|
The Cannabinoid Receptor Agonist WIN55,212-2 Ameliorates Hippocampal Neuronal Damage After Chronic Cerebral Hypoperfusion Possibly Through Inhibiting Oxidative Stress and ASK1-p38 Signaling. Neurotox Res 2019; 37:847-856. [PMID: 31808139 DOI: 10.1007/s12640-019-00141-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 11/10/2019] [Accepted: 11/14/2019] [Indexed: 12/16/2022]
Abstract
Chronic cerebral hypoperfusion (CCH) is a major contributor to cognitive decline and degenerative processes leading to Alzheimer's disease, vascular dementia, and aging. However, the delicate mechanism of CCH-induced neuronal damage, and therefore proper treatment, remains unclear. WIN55,212-2 (WIN) is a nonselective cannabinoid receptor agonist that has been shown to have effects on hippocampal neuron survival. In this study, we investigated the potential roles of WIN, as well as its underlying mechanism in a rat CCH model of bilateral common carotid artery occlusion. Hippocampal morphological changes and mitochondrial ultrastructure were detected using hematoxylin and eosin staining and electron microscopy, respectively. Various biomarkers, such as reactive oxidative species (ROS), superoxide dismutase (SOD), catalase (CAT), and malondialdehyde (MDA) were used to assess the level of oxidative stress in the hippocampus. Furthermore, the expression levels of neuronal nuclei (NeuN), apoptosis signal-regulating kinase 1 (ASK1)-p38 signaling proteins, cleaved Caspase-9 and -3, and cytochrome-c (Cyt-C) were accessed by western blotting. CCH decreased the levels of NeuN, Cyt-C (mitochondrial), SOD, and CAT, and increased the levels of MDA, phosphorylated ASK1 and phosphorylated p38, cleaved Caspase-9 and -3, and Cyt-C (cytoplasm), which were reversed by WIN treatment. Chronic treatment with WIN also improved CCH-induced neuronal degeneration and mitochondrial fragmentation. These findings indicated that WIN may be a potential therapeutic agent for ischemic neuronal damage, involving a mechanism associated with the suppression of oxidative stress and ASK1-p38 signaling.
Collapse
|
25
|
Bhatia P, Singh N. Ameliorative Effect of Phosphodiesterase-5 Inhibitor in Rat Model of Vascular Dementia. Curr Neurovasc Res 2019; 16:27-39. [DOI: 10.2174/1567202616666190130153954] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 01/03/2019] [Accepted: 01/05/2019] [Indexed: 12/15/2022]
Abstract
Introduction:
Cerebral hypoperfusion has been considered as major risk factor for Vascular
Dementia (VaD). The present study shows the potential of Tadalafil, a phosphodiesterase-5
inhibitor, in bilateral common carotid artery occlusion (BCCAo) induced VaD in rats.
Materials and Methods:
BCCAo procedure was performed under anesthesia in wistar rats to induce
VaD. Morris Water-Maze (MWM) parameter was employed on 7th day post-surgery to determine
learning and memory. Escape latency time, time spent in target quadrant, Path length and
average swim speed taken as important parameters in MWM. Endothelial dysfunction was assessed
in isolated aorta by observing endothelial dependent vasorelaxations and levels of serum
nitrite. Various biochemical and histopathological estimations were also performed.
Results:
BCCAo produced significant impairment in endothelium dependent vasorelaxation and a
decrease in serum nitrite levels indicating endothelial dysfunction. Further poor performance on
MWM represents impairment of learning and memory. There was a significant rise in brain oxidative
stress level (indicated by increase in brain thiobarbituric acid reactive species and decrease in
reduced glutathione levels). Increase in brain acetylcholinesterase activity; brain myloperoxidase
activity and brain neutrophil infiltration (as marker of inflammation) were also observed.
Treatment of Tadalafil (5 & 10 mg/kg, p. o.)/Donepezil (0. 5 mg/kg, i.p., serving as standard)
ameliorated BCCAo induced endothelial dysfunction; memory deficits; biochemical and
histopathological changes in a significant manner.
Conclusion:
It may be concluded that Tadalafil has shown efficacy in rat model of BCCAo induced
VaD and that phosphodiesterase-5 can be considered as an important therapeutic target for
the treatment of VaD.
Collapse
Affiliation(s)
- Pankaj Bhatia
- CNS Research Lab., Pharmacology Division, Department of Pharmaceutical Sciences and Drug Research, Faculty of Medicine, Punjabi University, Patiala - 147002, Punjab, India
| | - Nirmal Singh
- CNS Research Lab., Pharmacology Division, Department of Pharmaceutical Sciences and Drug Research, Faculty of Medicine, Punjabi University, Patiala - 147002, Punjab, India
| |
Collapse
|
26
|
Aldea R, Weller RO, Wilcock DM, Carare RO, Richardson G. Cerebrovascular Smooth Muscle Cells as the Drivers of Intramural Periarterial Drainage of the Brain. Front Aging Neurosci 2019; 11:1. [PMID: 30740048 PMCID: PMC6357927 DOI: 10.3389/fnagi.2019.00001] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 01/07/2019] [Indexed: 12/25/2022] Open
Abstract
The human brain is the organ with the highest metabolic activity but it lacks a traditional lymphatic system responsible for clearing waste products. We have demonstrated that the basement membranes of cerebral capillaries and arteries represent the lymphatic pathways of the brain along which intramural periarterial drainage (IPAD) of soluble metabolites occurs. Failure of IPAD could explain the vascular deposition of the amyloid-beta protein as cerebral amyloid angiopathy (CAA), which is a key pathological feature of Alzheimer's disease. The underlying mechanisms of IPAD, including its motive force, have not been clarified, delaying successful therapies for CAA. Although arterial pulsations from the heart were initially considered to be the motive force for IPAD, they are not strong enough for efficient IPAD. This study aims to unravel the driving force for IPAD, by shifting the perspective of a heart-driven clearance of soluble metabolites from the brain to an intrinsic mechanism of cerebral arteries (e.g., vasomotion-driven IPAD). We test the hypothesis that the cerebrovascular smooth muscle cells, whose cycles of contraction and relaxation generate vasomotion, are the drivers of IPAD. A novel multiscale model of arteries, in which we treat the basement membrane as a fluid-filled poroelastic medium deformed by the contractile cerebrovascular smooth muscle cells, is used to test the hypothesis. The vasomotion-induced intramural flow rates suggest that vasomotion-driven IPAD is the only mechanism postulated to date capable of explaining the available experimental observations. The cerebrovascular smooth muscle cells could represent valuable drug targets for prevention and early interventions in CAA.
Collapse
Affiliation(s)
- Roxana Aldea
- Mathematical Sciences, University of Southampton, Southampton, United Kingdom
| | - Roy O Weller
- Clinical Neurosciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
| | - Donna M Wilcock
- Department of Physiology, Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States
| | - Roxana O Carare
- Clinical Neurosciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, United Kingdom
| | - Giles Richardson
- Mathematical Sciences, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
27
|
Wang DP, Yin H, Kang K, Lin Q, Su SH, Hai J. The potential protective effects of cannabinoid receptor agonist WIN55,212-2 on cognitive dysfunction is associated with the suppression of autophagy and inflammation in an experimental model of vascular dementia. Psychiatry Res 2018; 267:281-288. [PMID: 29945070 DOI: 10.1016/j.psychres.2018.06.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 06/07/2018] [Accepted: 06/07/2018] [Indexed: 12/17/2022]
Abstract
Vascular dementia (VaD) is characteristic of chronic brain ischemia and progressive memory decline, which has a high incidence in the elderly. However, there are no effective treatments for VaD, and the underlying mechanism of its pathogenesis remains unclear. This study investigated the effects of a synthetic cannabinoid receptor agonist WIN55,212-2 (WIN) on VaD, and molecular mechanisms of the effects. VaD model was induced by 2-vessel occlusion (2VO). Spatial reference learning was evaluated by the Morris water maze, and recognition memory was assessed using the novel object recognition test. Autophagy-related proteins [microtubule-associated protein 1 light chain 3 (LC-3) and Beclin-1] were examined by immunohistochemistry and Western blot. Caspase-3 was detected by Western blot. Inflammatory factors, tumor necrosis factor alpha (TNF-α) and interleukin 1 beta (IL-1β), were estimated by reverse transcription-polymerase chain reaction (RT-PCR) and Western blot. VaD increased the levels of LC-3, Beclin-1, and inflammatory factors, which were reversed by chronic treatment with WIN. WIN decreased the expression of Capase-3, and improved the learning and memory impairment of VaD rats. These data indicate that WIN exerts a neuroprotective effect on the cognitive deficits of VaD rats, which may be associated with the suppression of excessive autophagy and inflammation.
Collapse
Affiliation(s)
- Da-Peng Wang
- Department of Neurosurgery, Tong Ji Hospital, Tong Ji University School of Medicine, Shanghai 200065, China
| | - Hang Yin
- Department of Neurosurgery, Zao Zhuang Municipal Hospital, Zaozhuang, Shandong 277000, China
| | - Kai Kang
- Department of Research and Surveillance Evaluation, Shanghai Center for Health Promotion, Shanghai 200040, China
| | - Qi Lin
- Department of Pharmacy, Institutes of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Shao-Hua Su
- Department of Neurosurgery, Tong Ji Hospital, Tong Ji University School of Medicine, Shanghai 200065, China.
| | - Jian Hai
- Department of Neurosurgery, Tong Ji Hospital, Tong Ji University School of Medicine, Shanghai 200065, China.
| |
Collapse
|
28
|
Tiwari N, Bhatia P, Kumar A, Jaggi AS, Singh N. Potential of carnosine, a histamine precursor in rat model of bilateral common carotid artery occlusion-induced vascular dementia. Fundam Clin Pharmacol 2018; 32:516-531. [DOI: 10.1111/fcp.12376] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 03/28/2018] [Accepted: 04/13/2018] [Indexed: 01/04/2023]
Affiliation(s)
- Nidhi Tiwari
- CNS Research Lab.; Pharmacology Division; Department of Pharmaceutical Sciences and Drug Research; Faculty of Medicine; Punjabi University; Patiala 147002 Punjab India
| | - Pankaj Bhatia
- CNS Research Lab.; Pharmacology Division; Department of Pharmaceutical Sciences and Drug Research; Faculty of Medicine; Punjabi University; Patiala 147002 Punjab India
| | - Amit Kumar
- CNS Research Lab.; Pharmacology Division; Department of Pharmaceutical Sciences and Drug Research; Faculty of Medicine; Punjabi University; Patiala 147002 Punjab India
- Pharmacology Division; Maharaja Agrasen School of Pharmacy; Maharaja Agrasen University; Baddi 174103 Himachal Pradesh India
| | - Amteshwar S. Jaggi
- Pharmacology Division; Department of Pharmaceutical Sciences and Drug Research; Faculty of Medicine; Punjabi University; Patiala 147002 Punjab India
| | - Nirmal Singh
- Pharmacology Division; Department of Pharmaceutical Sciences and Drug Research; Faculty of Medicine; Punjabi University; Patiala 147002 Punjab India
| |
Collapse
|
29
|
Vinters HV, Zarow C, Borys E, Whitman JD, Tung S, Ellis WG, Zheng L, Chui HC. Review: Vascular dementia: clinicopathologic and genetic considerations. Neuropathol Appl Neurobiol 2018; 44:247-266. [DOI: 10.1111/nan.12472] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 01/13/2018] [Indexed: 12/21/2022]
Affiliation(s)
- H. V. Vinters
- Departments of Pathology & Laboratory Medicine (Neuropathology) and Neurology; David Geffen School of Medicine at UCLA; Los Angeles CA USA
| | - C. Zarow
- Department of Neurology; Keck School of Medicine at University of Southern California; Los Angeles CA USA
| | - E. Borys
- Department of Pathology; University of California Davis School of Medicine; Sacramento CA USA
- Department of Pathology; Loyola University Medical Center; Maywood IL USA
| | - J. D. Whitman
- Departments of Pathology & Laboratory Medicine (Neuropathology) and Neurology; David Geffen School of Medicine at UCLA; Los Angeles CA USA
- Departments of Pathology & Laboratory Medicine; UC San Francisco Medical Center; San Francisco CA USA
| | - S. Tung
- Departments of Pathology & Laboratory Medicine (Neuropathology) and Neurology; David Geffen School of Medicine at UCLA; Los Angeles CA USA
| | - W. G. Ellis
- Department of Pathology; University of California Davis School of Medicine; Sacramento CA USA
| | - L. Zheng
- Department of Neurology; Keck School of Medicine at University of Southern California; Los Angeles CA USA
| | - H. C. Chui
- Department of Neurology; Keck School of Medicine at University of Southern California; Los Angeles CA USA
| |
Collapse
|
30
|
Tuttolomondo A, Petta S, Casuccio A, Maida C, Corte VD, Daidone M, Di Raimondo D, Pecoraro R, Fonte R, Cirrincione A, Zafonte R, Cabibi D, Cammà C, Di Marco V, Licata A, Magliozzo F, Marchesini G, Merlino G, Craxì A, Pinto A. Reactive hyperemia index (RHI) and cognitive performance indexes are associated with histologic markers of liver disease in subjects with non-alcoholic fatty liver disease (NAFLD): a case control study. Cardiovasc Diabetol 2018; 17:28. [PMID: 29452601 PMCID: PMC5815178 DOI: 10.1186/s12933-018-0670-7] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 02/01/2018] [Indexed: 12/13/2022] Open
Abstract
Background No study evaluated vascular health markers in subjects with non-alcoholic fatty liver disease (NAFLD) through a combined analysis of reactive hyperemia peripheral arterial tonometry (RH-PAT) and arterial stiffness indexes. Aim of the study We aimed to assess whether NAFLD and its histological severity are associated with impairment of arterial stiffness and RH-PAT indexes in a mixed cohort of patients with biopsy-proven NAFLD. Materials and methods The Kleiner classification was used to grade NAFLD grade. Pulse wave velocity (PWV) and augmentation index (Aix) were used as markers of arterial stiffness, whereas endothelial function was assessed using reactive hyperemia index (RHI). The mini-mental state examination (MMSE) was administered to test cognitive performance. Results 80 consecutive patients with biopsy-proven NAFLD and 83 controls without fatty liver disease. NAFLD subjects showed significantly lower mean RHI, higher mean arterial stiffness indexes and lower mean MMSE score. Multivariable analysis after correction for BMI, dyslipidaemia, hypertension, sex, diabetes, age and cardiovascular disease showed that BMI, diastolic blood pressure and RHI are significantly associated to NAFLD. Simple linear regression analysis showed among non-alcoholic steatohepatitis (NASH) subjects a significant negative relationship between ballooning grade and MMSE and a significant positive association between Kleiner steatosis grade and augmentation index. Conclusions Future research will be addressed to evaluate the relationship between inflammatory markers and arterial stiffness and endothelial function indexes in NAFLD subjects. These study will evaluate association between cardiovascular event incidence and arterial stiffness, endothelial and cognitive markers, and they will address the beneficial effects of cardiovascular drugs such as statins and ACE inhibitors on these surrogate markers in NAFLD subjects.
Collapse
Affiliation(s)
- Antonino Tuttolomondo
- U.O.C di Medicina Interna con Stroke Care, Dipartimento Biomedico di Medicina Interna e Specialistica (Di.Bi.M.I.S), University of Palermo, P.zza delle Cliniche n.2, Palermo, 90127, Italy.
| | - Salvatore Petta
- Sezione di Gastroenterologia e Epatologia, Dipartimento Biomedico di Medicina Interna e Specialistica (Di.Bi.M.I.S), University of Palermo, Palermo, Italy
| | - Alessandra Casuccio
- Dipartimento di Scienze per la Promozione della Salute e Materno Infantile, University of Palermo, Palermo, Italy
| | - Carlo Maida
- U.O.C di Medicina Interna con Stroke Care, Dipartimento Biomedico di Medicina Interna e Specialistica (Di.Bi.M.I.S), University of Palermo, P.zza delle Cliniche n.2, Palermo, 90127, Italy
| | - Vittoriano Della Corte
- U.O.C di Medicina Interna con Stroke Care, Dipartimento Biomedico di Medicina Interna e Specialistica (Di.Bi.M.I.S), University of Palermo, P.zza delle Cliniche n.2, Palermo, 90127, Italy
| | - Mario Daidone
- U.O.C di Medicina Interna con Stroke Care, Dipartimento Biomedico di Medicina Interna e Specialistica (Di.Bi.M.I.S), University of Palermo, P.zza delle Cliniche n.2, Palermo, 90127, Italy
| | - Domenico Di Raimondo
- U.O.C di Medicina Interna con Stroke Care, Dipartimento Biomedico di Medicina Interna e Specialistica (Di.Bi.M.I.S), University of Palermo, P.zza delle Cliniche n.2, Palermo, 90127, Italy
| | - Rosaria Pecoraro
- U.O.C di Medicina Interna con Stroke Care, Dipartimento Biomedico di Medicina Interna e Specialistica (Di.Bi.M.I.S), University of Palermo, P.zza delle Cliniche n.2, Palermo, 90127, Italy
| | - Roberto Fonte
- U.O.C di Medicina Interna con Stroke Care, Dipartimento Biomedico di Medicina Interna e Specialistica (Di.Bi.M.I.S), University of Palermo, P.zza delle Cliniche n.2, Palermo, 90127, Italy
| | - Anna Cirrincione
- U.O.C di Medicina Interna con Stroke Care, Dipartimento Biomedico di Medicina Interna e Specialistica (Di.Bi.M.I.S), University of Palermo, P.zza delle Cliniche n.2, Palermo, 90127, Italy
| | | | - Daniela Cabibi
- Cattedra di Anatomia Patologica, University of Palermo, Palermo, Italy
| | - Calogero Cammà
- Sezione di Gastroenterologia e Epatologia, Dipartimento Biomedico di Medicina Interna e Specialistica (Di.Bi.M.I.S), University of Palermo, Palermo, Italy
| | - Vito Di Marco
- Sezione di Gastroenterologia e Epatologia, Dipartimento Biomedico di Medicina Interna e Specialistica (Di.Bi.M.I.S), University of Palermo, Palermo, Italy
| | - Anna Licata
- Sezione di Gastroenterologia e Epatologia, Dipartimento Biomedico di Medicina Interna e Specialistica (Di.Bi.M.I.S), University of Palermo, Palermo, Italy
| | | | - Giulio Marchesini
- Dipartimento di Scienze Mediche e Chirurgiche, "Alma Mater Studiorum", University of Bologna, Bologna, Italy
| | | | - Antonio Craxì
- Sezione di Gastroenterologia e Epatologia, Dipartimento Biomedico di Medicina Interna e Specialistica (Di.Bi.M.I.S), University of Palermo, Palermo, Italy
| | - Antonio Pinto
- U.O.C di Medicina Interna con Stroke Care, Dipartimento Biomedico di Medicina Interna e Specialistica (Di.Bi.M.I.S), University of Palermo, P.zza delle Cliniche n.2, Palermo, 90127, Italy
| |
Collapse
|
31
|
Hu M, Liu Z, Lv P, Wang H, Zhu Y, Qi Q, Xu J, Gao L. Nimodipine activates neuroprotective signaling events and inactivates autophages in the VCID rat hippocampus. Neurol Res 2017; 39:904-909. [PMID: 28782464 DOI: 10.1080/01616412.2017.1356157] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND Autophagy and phosphatidylinositol 3-kinase (PI3K)/Akt kinase pathways are implicated in cognitive decline associated with cerebrovascular lesions. This decline is reflected in the concept of vascular cognitive impairment and dementia (VCID). However, the underlying molecular mechanism and specific details regarding these types of cognitive deficits induced by chronic brain hypoperfusion have not been elucidated. METHODS We designed a method to evaluate these mechanisms. Adult male Sprague-Dawley rats were subjected to permanent bilateral occlusion of the common carotid artery (2VO) and randomly divided into three groups: Sham, Vehicle (2VO), and Nimodipine10 (2VO + nimodipine 10 mg/kg). Each group was studied for 4 weeks postoperatively and assessed by the Morris water maze. RESULTS The results of this study show that chronic brain hypoperfusion significantly increased the number of autophagic vacuoles with high LC3 II levels, but it decreased p-Akt and p-CREB levels, which were involved in the PI3K/Akt kinase pathway in the hippocampi of rats. Additionally, significant cognitive losses were observed following 2VO. Further analysis showed that, in VCID rats subjected to 2VO, nimodipine administration decreased autophagy, increased the Akt/CREB signaling pathway and significantly reduced brain damage. CONCLUSIONS We concluded that neuronal pathology and activation of the autophagic and Akt/CREB signaling pathway caused by chronic brain hypoperfusion could suppress cognitive behavior, which may provide a novel way for the prevention of VCID. The results of this study indicate that nimodipine protected the brain from chronic brain hypoperfusion damage by suppressing autophagy and activating the Akt/CREB signaling pathway.
Collapse
Affiliation(s)
- Ming Hu
- a Department of Neurology , Hebei General Hospital , Shijiazhuang , People's Republic of China
| | - Zhijuan Liu
- a Department of Neurology , Hebei General Hospital , Shijiazhuang , People's Republic of China
| | - Peiyuan Lv
- a Department of Neurology , Hebei General Hospital , Shijiazhuang , People's Republic of China
| | - Hebo Wang
- a Department of Neurology , Hebei General Hospital , Shijiazhuang , People's Republic of China
| | - Yifei Zhu
- b Department of Neurology , the Second Hospital of Hebei Medical University , Shijiazhuang , People's Republic of China
| | - Qianqian Qi
- a Department of Neurology , Hebei General Hospital , Shijiazhuang , People's Republic of China
| | - Jing Xu
- a Department of Neurology , Hebei General Hospital , Shijiazhuang , People's Republic of China
| | - Lei Gao
- c Department of Ultrasonography , the First Central Hospital of Baoding , Baoding , People's Republic of China
| |
Collapse
|
32
|
Smith EE, Cieslak A, Barber P, Chen J, Chen YW, Donnini I, Edwards JD, Frayne R, Field TS, Hegedus J, Hanganu V, Ismail Z, Kanji J, Nakajima M, Noor R, Peca S, Sahlas D, Sharma M, Sposato LA, Swartz RH, Zerna C, Black SE, Hachinski V. Therapeutic Strategies and Drug Development for Vascular Cognitive Impairment. J Am Heart Assoc 2017; 6:e005568. [PMID: 28476873 PMCID: PMC5524100 DOI: 10.1161/jaha.117.005568] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
| | | | | | | | - Yu-Wei Chen
- National Taiwan University Hospital, Taipei, Taiwan
- Taiwan Landseed Hospital, Taoyuan, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Meissner A, Minnerup J, Soria G, Planas AM. Structural and functional brain alterations in a murine model of Angiotensin II-induced hypertension. J Neurochem 2016; 140:509-521. [DOI: 10.1111/jnc.13905] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 11/07/2016] [Accepted: 11/09/2016] [Indexed: 01/18/2023]
Affiliation(s)
- Anja Meissner
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS); Barcelona Spain
- Department of Neurology; University Hospital Münster; Münster Germany
| | - Jens Minnerup
- Department of Neurology; University Hospital Münster; Münster Germany
| | - Guadalupe Soria
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS); Barcelona Spain
| | - Anna M Planas
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS); Barcelona Spain
- Departament d'Isquèmia Cerebral i Neurodegeneració; Institut d'Investigacions Biomèdiques de Barcelona (IIBB); Consejo Superior de Investigaciones Científicas (CSIC); Barcelona Spain
| |
Collapse
|