1
|
Carnevale L, Lembo G. Imaging the cerebral vasculature at different scales: translational tools to investigate the neurovascular interfaces. Cardiovasc Res 2025; 120:2373-2384. [PMID: 39082279 DOI: 10.1093/cvr/cvae165] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 04/26/2024] [Accepted: 05/23/2024] [Indexed: 04/09/2025] Open
Abstract
The improvements in imaging technology opened up the possibility to investigate the structure and function of cerebral vasculature and the neurovascular unit with unprecedented precision and gaining deep insights not only on the morphology of the vessels but also regarding their function and regulation related to the cerebral activity. In this review, we will dissect the different imaging capabilities regarding the cerebrovascular tree, the neurovascular unit, the haemodynamic response function, and thus, the vascular-neuronal coupling. We will discuss both clinical and preclinical setting, with a final discussion on the current scenery in cerebrovascular imaging where magnetic resonance imaging and multimodal microscopy emerge as the most potent and versatile tools, respectively, in the clinical and preclinical context.
Collapse
Affiliation(s)
- Lorenzo Carnevale
- Department of AngioCardioNeurology and Translational Medicine, I.R.C.C.S. INM Neuromed, Via dell'Elettronica, 86077 Pozzilli, IS, Italy
| | - Giuseppe Lembo
- Department of AngioCardioNeurology and Translational Medicine, I.R.C.C.S. INM Neuromed, Via dell'Elettronica, 86077 Pozzilli, IS, Italy
- Department of Molecular Medicine, 'Sapienza' University of Rome, Viale Regina Elena, 291, 00161 Rome, Italy
| |
Collapse
|
2
|
Alves L, Hashiguchi D, Loss CM, van Praag H, Longo BM. Vascular dysfunction in Alzheimer's disease: Exploring the potential of aerobic and resistance exercises as therapeutic strategies. J Alzheimers Dis 2025; 104:963-979. [PMID: 40079781 DOI: 10.1177/13872877251321118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/15/2025]
Abstract
Alzheimer's disease (AD) is the leading cause of morbidity and mortality worldwide, as a result of cognitive decline and neurological dysfunction. In AD, reduced cerebral blood flow and impaired vascularization result from capillary bed degeneration and decreased angiogenesis, as observed in both patients and animal models. Physical exercise is recognized as a potential intervention to delay AD progression and reduce disease risk. While most studies have focused on the benefits of aerobic exercise (AE), emerging evidence suggests that resistance exercise (RE) also exerts positive effects on overall health and cognitive function in aging and AD. However, a notable gap in knowledge remains regarding the effects of RE on cerebral blood flow and vascular structure. This review explores the processes by which AE and RE influence brain vascularization in aging and AD, including blood flow, endothelial function, angiogenesis and neurotrophic factor levels. Based on pre-clinical and clinical studies, we conclude that both AE and RE contribute to improved cerebral blood flow and vascular function, promoting vascular repair in the aging and AD-affected brain. By examining the relationship between exercise modalities and brain vascularization, this review expands knowledge regarding the processes underlying the neuroprotective effects of exercise in neurodegenerative and aging conditions.
Collapse
Affiliation(s)
- Larissa Alves
- Departamento de Fisiologia, Universidade Federal de São Paulo, UNIFESP, São Paulo, SP, Brasil
| | - Debora Hashiguchi
- Instituto do Cérebro, Universidade Federal do Rio Grande do Norte, UFRN, Natal, RN, Brasil
| | - Cássio Morais Loss
- Stiles-Nicholson Brain Institute, Charles E. Schmidt College of Medicine, Florida Atlantic University, FAU, Jupiter, FL, USA
| | - Henriette van Praag
- Stiles-Nicholson Brain Institute, Charles E. Schmidt College of Medicine, Florida Atlantic University, FAU, Jupiter, FL, USA
| | - Beatriz Monteiro Longo
- Departamento de Fisiologia, Universidade Federal de São Paulo, UNIFESP, São Paulo, SP, Brasil
| |
Collapse
|
3
|
Lohkamp KJ, Timmer N, Solé Guardia G, Shenk J, Verweij V, Geenen B, Dederen PJ, Bakker L, Egitimci C, Yoldas R, Verhaeg M, Kothuis J, Nieuwenhuis D, Wiesmann M, Kiliaan AJ. Sex-Specific Adaptations in Alzheimer's Disease and Ischemic Stroke: A Longitudinal Study in Male and Female APP swe/PS1 dE9 Mice. Life (Basel) 2025; 15:333. [PMID: 40141679 PMCID: PMC11944048 DOI: 10.3390/life15030333] [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: 02/03/2025] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 03/28/2025] Open
Abstract
The long-term impact of stroke on Alzheimer's disease (AD) progression, particularly regarding sex-specific differences, remains unknown. Using a longitudinal study design, we investigated transient middle cerebral artery occlusion in 3.5-month-old APPswe/PS1dE9 (APP/PS1) and wild-type mice. In vivo, we assessed behavior, cerebral blood flow (CBF), and structural integrity by neuroimaging, as well as post-mortem myelin integrity (polarized light imaging, PLI), neuroinflammation, and amyloid beta (Aβ) deposition. APP/PS1 mice exhibited cognitive decline, white matter degeneration (reduced fractional anisotropy (FA) via diffusion tensor imaging (DTI)), and decreased myelin density via PLI. Despite early hypertension, APP/PS1 mice showed only sporadic hypoperfusion. Cortical thickening and hippocampal hypertrophy likely resulted from Aβ accumulation and neuroinflammation. Stroke-operated mice retained cognition despite cortical thinning and hippocampal atrophy due to cerebrovascular adaptation, including increased CBF in the hippocampus and thalamus. Stroke did not worsen AD pathology, nor did AD exacerbate stroke outcomes. Sex differences were found: female APP/PS1 mice had more severe Aβ deposition, hyperactivity, lower body weight, and reduced CBF but less neuroinflammation, suggesting potential neuroprotection. These findings highlight white matter degeneration and Aβ pathology as key drivers of cognitive decline in AD, with stroke-related deficits mitigated by (cerebro)vascular adaptation. Sex-specific therapies are crucial for AD and stroke.
Collapse
Affiliation(s)
- Klara J. Lohkamp
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, The Netherlands; (K.J.L.); (N.T.); (G.S.G.); (J.S.); (V.V.); (B.G.); (P.J.D.); (L.B.); (C.E.); (R.Y.); (M.V.); (J.K.); (M.W.)
| | - Nienke Timmer
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, The Netherlands; (K.J.L.); (N.T.); (G.S.G.); (J.S.); (V.V.); (B.G.); (P.J.D.); (L.B.); (C.E.); (R.Y.); (M.V.); (J.K.); (M.W.)
| | - Gemma Solé Guardia
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, The Netherlands; (K.J.L.); (N.T.); (G.S.G.); (J.S.); (V.V.); (B.G.); (P.J.D.); (L.B.); (C.E.); (R.Y.); (M.V.); (J.K.); (M.W.)
| | - Justin Shenk
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, The Netherlands; (K.J.L.); (N.T.); (G.S.G.); (J.S.); (V.V.); (B.G.); (P.J.D.); (L.B.); (C.E.); (R.Y.); (M.V.); (J.K.); (M.W.)
| | - Vivienne Verweij
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, The Netherlands; (K.J.L.); (N.T.); (G.S.G.); (J.S.); (V.V.); (B.G.); (P.J.D.); (L.B.); (C.E.); (R.Y.); (M.V.); (J.K.); (M.W.)
| | - Bram Geenen
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, The Netherlands; (K.J.L.); (N.T.); (G.S.G.); (J.S.); (V.V.); (B.G.); (P.J.D.); (L.B.); (C.E.); (R.Y.); (M.V.); (J.K.); (M.W.)
| | - Pieter J. Dederen
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, The Netherlands; (K.J.L.); (N.T.); (G.S.G.); (J.S.); (V.V.); (B.G.); (P.J.D.); (L.B.); (C.E.); (R.Y.); (M.V.); (J.K.); (M.W.)
| | - Lieke Bakker
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, The Netherlands; (K.J.L.); (N.T.); (G.S.G.); (J.S.); (V.V.); (B.G.); (P.J.D.); (L.B.); (C.E.); (R.Y.); (M.V.); (J.K.); (M.W.)
- Alzheimer Center Limburg, Department of Psychiatry and Neuropsychology, Mental Health and Neuroscience Research Institute (MHeNs), European Graduate School of Neuroscience (EURON), Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Cansu Egitimci
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, The Netherlands; (K.J.L.); (N.T.); (G.S.G.); (J.S.); (V.V.); (B.G.); (P.J.D.); (L.B.); (C.E.); (R.Y.); (M.V.); (J.K.); (M.W.)
| | - Rengin Yoldas
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, The Netherlands; (K.J.L.); (N.T.); (G.S.G.); (J.S.); (V.V.); (B.G.); (P.J.D.); (L.B.); (C.E.); (R.Y.); (M.V.); (J.K.); (M.W.)
| | - Minou Verhaeg
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, The Netherlands; (K.J.L.); (N.T.); (G.S.G.); (J.S.); (V.V.); (B.G.); (P.J.D.); (L.B.); (C.E.); (R.Y.); (M.V.); (J.K.); (M.W.)
| | - Josine Kothuis
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, The Netherlands; (K.J.L.); (N.T.); (G.S.G.); (J.S.); (V.V.); (B.G.); (P.J.D.); (L.B.); (C.E.); (R.Y.); (M.V.); (J.K.); (M.W.)
| | - Desirée Nieuwenhuis
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, The Netherlands; (K.J.L.); (N.T.); (G.S.G.); (J.S.); (V.V.); (B.G.); (P.J.D.); (L.B.); (C.E.); (R.Y.); (M.V.); (J.K.); (M.W.)
| | - Maximilian Wiesmann
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, The Netherlands; (K.J.L.); (N.T.); (G.S.G.); (J.S.); (V.V.); (B.G.); (P.J.D.); (L.B.); (C.E.); (R.Y.); (M.V.); (J.K.); (M.W.)
| | - Amanda J. Kiliaan
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, The Netherlands; (K.J.L.); (N.T.); (G.S.G.); (J.S.); (V.V.); (B.G.); (P.J.D.); (L.B.); (C.E.); (R.Y.); (M.V.); (J.K.); (M.W.)
| |
Collapse
|
4
|
Qin H, Suo S, Yang F, Hao P, Zhang X. The role of digestive system diseases in cerebrovascular disease: a comprehensive Mendelian randomization study. Front Neurol 2024; 15:1389352. [PMID: 38854966 PMCID: PMC11157012 DOI: 10.3389/fneur.2024.1389352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 05/13/2024] [Indexed: 06/11/2024] Open
Abstract
Background Cerebrovascular disease, among the most prevalent neurological disorders, poses a substantial threat to human health with its elevated mortality and disability rates, placing considerable strain on healthcare systems. Although several studies in recent years have suggested a potential association between digestive system diseases and cerebrovascular diseases, the findings remain inconsistent. Methods Genome-wide association study (GWAS) summary data for 12 digestive diseases and cerebrovascular diseases were used to conduct Mendelian randomization (MR) analysis. In this investigation, we endeavored to elucidate the causal relationship between digestive system diseases and cerebrovascular diseases. Employing a comprehensive approach, including two-sample MR (TSMR), multivariate MR (MVMR), and two-step MR analysis, we leveraged summary statistics data obtained from published GWAS. The primary analysis method employed was inverse variance weighted (IVW), with MR-Egger and weighted median (WM) as secondary methods. Sensitivity analysis included heterogeneity testing, horizontal multivariate testing, MR-PRESSO, and a "leave-one-out" method. Additionally, the F-statistic was utilized to assess the strength of instrumental variables, ensuring robust results. Results In the TSMR analysis, this study found a significant causal relationship between genetically predicted gastroesophageal reflux disease (GERD) and any stroke (AS), any ischemic stroke (AIS), large-artery atherosclerotic stroke (LAS), intracranial aneurysm (IA), and subarachnoid hemorrhage (SAH). In MVMR analysis, this study found that even after adjusting for systolic blood pressure (SBP), body mass index (BMI) and type 2 diabetes (T2D), the causal relationship remains exist. In the two-step MR mediation analysis, it was found that BMI, SBP and T2D play mediating role in the causal relationship between GERD and cerebrovascular diseases. Conclusion This study indicates a clear positive causal relationship between GERD and cerebrovascular diseases, and this causal association remains significant even after adjusting for BMI, SBP and T2D. The mediation MR analysis suggests that BMI, SBP and T2D may mediate the causal relationship between GERD and the risk of cerebrovascular diseases.
Collapse
Affiliation(s)
- Hao Qin
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| | - Shihuan Suo
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| | - Fan Yang
- Department of Anesthesiology, The First Hospital of Jilin University, Changchun, China
| | - Pengfei Hao
- Department of Neurosurgery, Tongji Shanxi Hospital, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Taiyuan, China
| | - Xianfeng Zhang
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
5
|
Zhang M, Zhang Z, Li H, Xia Y, Xing M, Xiao C, Cai W, Bu L, Li Y, Park TE, Tang Y, Ye X, Lin WJ. Blockage of VEGF function by bevacizumab alleviates early-stage cerebrovascular dysfunction and improves cognitive function in a mouse model of Alzheimer's disease. Transl Neurodegener 2024; 13:1. [PMID: 38173017 PMCID: PMC10763201 DOI: 10.1186/s40035-023-00388-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 11/14/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurodegenerative disorder and the predominant type of dementia worldwide. It is characterized by the progressive and irreversible decline of cognitive functions. In addition to the pathological beta-amyloid (Aβ) deposition, glial activation, and neuronal injury in the postmortem brains of AD patients, increasing evidence suggests that the often overlooked vascular dysfunction is an important early event in AD pathophysiology. Vascular endothelial growth factor (VEGF) plays a critical role in regulating physiological functions and pathological changes in blood vessels, but whether VEGF is involved in the early stage of vascular pathology in AD remains unclear. METHODS We used an antiangiogenic agent for clinical cancer treatment, the humanized monoclonal anti-VEGF antibody bevacizumab, to block VEGF binding to its receptors in the 5×FAD mouse model at an early age. After treatment, memory performance was evaluated by a novel object recognition test, and cerebral vascular permeability and perfusion were examined by an Evans blue assay and blood flow scanning imaging analysis. Immunofluorescence staining was used to measure glial activation and Aβ deposits. VEGF and its receptors were analyzed by enzyme-linked immunosorbent assay and immunoblotting. RNA sequencing was performed to elucidate bevacizumab-associated transcriptional signatures in the hippocampus of 5×FAD mice. RESULTS Bevacizumab treatment administered from 4 months of age dramatically improved cerebrovascular functions, reduced glial activation, and restored long-term memory in both sexes of 5×FAD mice. Notably, a sex-specific change in different VEGF receptors was identified in the cortex and hippocampus of 5×FAD mice. Soluble VEGFR1 was decreased in female mice, while full-length VEGFR2 was increased in male mice. Bevacizumab treatment reversed the altered expression of receptors to be comparable to the level in the wild-type mice. Gene Set Enrichment Analysis of transcriptomic changes revealed that bevacizumab effectively reversed the changes in the gene sets associated with blood-brain barrier integrity and vascular smooth muscle contraction in 5×FAD mice. CONCLUSIONS Our study demonstrated the mechanistic roles of VEGF at the early stage of amyloidopathy and the protective effects of bevacizumab on cerebrovascular function and memory performance in 5×FAD mice. These findings also suggest the therapeutic potential of bevacizumab for the early intervention of AD.
Collapse
Affiliation(s)
- Min Zhang
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510120, China
| | - Zhan Zhang
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510120, China
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan, 528200, China
| | - Honghong Li
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Yuting Xia
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan, 528200, China
| | - Mengdan Xing
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan, 528200, China
| | - Chuan Xiao
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan, 528200, China
| | - Wenbao Cai
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510120, China
| | - Lulu Bu
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Yi Li
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Tae-Eun Park
- Department of Biomedical Engineering, College of Information and Biotechnology, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Yamei Tang
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510120, China.
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan, 528200, China.
| | - Xiaojing Ye
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510120, China.
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510120, China.
| | - Wei-Jye Lin
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan, 528200, China.
| |
Collapse
|
6
|
Epp R, Glück C, Binder NF, El Amki M, Weber B, Wegener S, Jenny P, Schmid F. The role of leptomeningeal collaterals in redistributing blood flow during stroke. PLoS Comput Biol 2023; 19:e1011496. [PMID: 37871109 PMCID: PMC10621965 DOI: 10.1371/journal.pcbi.1011496] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 11/02/2023] [Accepted: 09/03/2023] [Indexed: 10/25/2023] Open
Abstract
Leptomeningeal collaterals (LMCs) connect the main cerebral arteries and provide alternative pathways for blood flow during ischaemic stroke. This is beneficial for reducing infarct size and reperfusion success after treatment. However, a better understanding of how LMCs affect blood flow distribution is indispensable to improve therapeutic strategies. Here, we present a novel in silico approach that incorporates case-specific in vivo data into a computational model to simulate blood flow in large semi-realistic microvascular networks from two different mouse strains, characterised by having many and almost no LMCs between middle and anterior cerebral artery (MCA, ACA) territories. This framework is unique because our simulations are directly aligned with in vivo data. Moreover, it allows us to analyse perfusion characteristics quantitatively across all vessel types and for networks with no, few and many LMCs. We show that the occlusion of the MCA directly caused a redistribution of blood that was characterised by increased flow in LMCs. Interestingly, the improved perfusion of MCA-sided microvessels after dilating LMCs came at the cost of a reduced blood supply in other brain areas. This effect was enhanced in regions close to the watershed line and when the number of LMCs was increased. Additional dilations of surface and penetrating arteries after stroke improved perfusion across the entire vasculature and partially recovered flow in the obstructed region, especially in networks with many LMCs, which further underlines the role of LMCs during stroke.
Collapse
Affiliation(s)
- Robert Epp
- Institute of Fluid Dynamics, ETH Zurich, Zurich, Switzerland
| | - Chaim Glück
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Nadine Felizitas Binder
- Deptartment of Neurology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Mohamad El Amki
- Deptartment of Neurology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Bruno Weber
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Susanne Wegener
- Deptartment of Neurology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Patrick Jenny
- Institute of Fluid Dynamics, ETH Zurich, Zurich, Switzerland
| | - Franca Schmid
- Institute of Fluid Dynamics, ETH Zurich, Zurich, Switzerland
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- ARTORG Center for Biomedical Engineering Research, University of Bern, Bern, Switzerland
| |
Collapse
|
7
|
Chen HF, Lambers H, Nagelmann N, Sandbrink M, Segelcke D, Pogatzki-Zahn E, Faber C, Pradier B. Generation of a whole-brain hemodynamic response function and sex-specific differences in cerebral processing of mechano-sensation in mice detected by BOLD fMRI. Front Neurosci 2023; 17:1187328. [PMID: 37700753 PMCID: PMC10493293 DOI: 10.3389/fnins.2023.1187328] [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: 03/15/2023] [Accepted: 07/05/2023] [Indexed: 09/14/2023] Open
Abstract
BOLD fMRI has become a prevalent method to study cerebral sensory processing in rodent disease models, including pain and mechanical hypersensitivity. fMRI data analysis is frequently combined with a general-linear-model (GLM) -based analysis, which uses the convolution of a hemodynamic response function (HRF) with the stimulus paradigm. However, several studies indicated that the HRF differs across species, sexes, brain structures, and experimental factors, including stimulation modalities or anesthesia, and hence might strongly affect the outcome of BOLD analyzes. While considerable work has been done in humans and rats to understand the HRF, much less is known in mice. As a prerequisite to investigate mechano-sensory processing and BOLD fMRI data in male and female mice, we (1) designed a rotating stimulator that allows application of two different mechanical modalities, including innocuous von Frey and noxious pinprick stimuli and (2) determined and statistically compared HRFs across 30 brain structures and experimental conditions, including sex and, stimulus modalities. We found that mechanical stimulation lead to brain-wide BOLD signal changes thereby allowing extraction of HRFs from multiple brain structures. However, we did not find differences in HRFs across all brain structures and experimental conditions. Hence, we computed a whole-brain mouse HRF, which is based on 88 functional scans from 30 mice. A comparison of this mouse-specific HRF with our previously reported rat-derived HRF showed significantly slower kinetics in mice. Finally, we detected pronounced differences in cerebral BOLD activation between male and female mice with mechanical stimulation, thereby exposing divergent processing of noxious and innocuous stimuli in both sexes.
Collapse
Affiliation(s)
- Hui-Fen Chen
- Clinic of Radiology, Translational Research Imaging Center (TRIC), University of Münster, Münster, Germany
| | - Henriette Lambers
- Clinic of Radiology, Translational Research Imaging Center (TRIC), University of Münster, Münster, Germany
| | - Nina Nagelmann
- Clinic of Radiology, Translational Research Imaging Center (TRIC), University of Münster, Münster, Germany
| | - Martin Sandbrink
- Clinic of Radiology, Translational Research Imaging Center (TRIC), University of Münster, Münster, Germany
| | - Daniel Segelcke
- Department of Anesthesiology, Intensive Care and Pain Medicine, University of Münster, Münster, Germany
| | - Esther Pogatzki-Zahn
- Department of Anesthesiology, Intensive Care and Pain Medicine, University of Münster, Münster, Germany
| | - Cornelius Faber
- Clinic of Radiology, Translational Research Imaging Center (TRIC), University of Münster, Münster, Germany
| | - Bruno Pradier
- Clinic of Radiology, Translational Research Imaging Center (TRIC), University of Münster, Münster, Germany
- Department of Anesthesiology, Intensive Care and Pain Medicine, University of Münster, Münster, Germany
| |
Collapse
|
8
|
Ruiz-Uribe NE, Bracko O, Swallow M, Omurzakov A, Dash S, Uchida H, Xiang D, Haft-Javaherian M, Falkenhain K, Lamont ME, Ali M, Njiru BN, Chang HY, Tan AY, Xiang JZ, Iadecola C, Park L, Sanchez T, Nishimura N, Schaffer CB. Vascular oxidative stress causes neutrophil arrest in brain capillaries, leading to decreased cerebral blood flow and contributing to memory impairment in a mouse model of Alzheimer’s disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.15.528710. [PMID: 36824768 PMCID: PMC9949082 DOI: 10.1101/2023.02.15.528710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
INTRODUCTION In this study, we explore the role of oxidative stress produced by NOX2-containing NADPH oxidase as a molecular mechanism causing capillary stalling and cerebral blood flow deficits in the APP/PS1 mouse model of AD. METHODS We inhibited NOX2 in APP/PS1 mice by administering a 10 mg/kg dose of the peptide inhibitor gp91-ds-tat i.p., for two weeks. We used in vivo two-photon imaging to measure capillary stalling, penetrating arteriole flow, and vascular inflammation. We also characterized short-term memory function and gene expression changes in cerebral microvessels. RESULTS We found that after NOX2 inhibition capillary stalling, as well as parenchymal and vascular inflammation, were significantly reduced. In addition, we found a significant increase in penetrating arteriole flow, followed by an improvement in short-term memory, and downregulation of inflammatory gene expression pathways. DISCUSSION Oxidative stress is a major mechanism leading to microvascular dysfunction in AD, and represents an important therapeutic target.
Collapse
|
9
|
Yang Y, Zhu Q, Wang L, Gao D, Wang Z, Geng Z. Effects of hypertension and aging on brain function in spontaneously hypertensive rats: a longitudinal resting-state functional magnetic resonance imaging study. Cereb Cortex 2022; 33:5493-5500. [PMID: 36408643 PMCID: PMC10152091 DOI: 10.1093/cercor/bhac436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/07/2022] [Accepted: 10/09/2022] [Indexed: 11/22/2022] Open
Abstract
Abstract
To investigate the dynamic evolution of brain function under the comorbidities of hypertension and aging. Resting-state functional magnetic resonance imaging scans were longitudinally acquired at 10, 24, and 52 weeks in spontaneously hypertensive rats (SHRs) and Wistar-Kyoto rats. We computed the mean amplitude of low-frequency fluctuation (mALFF), mean regional homogeneity (mReHo), and functional connectivity (FC). There was no interaction between hypertension and aging on brain function. The main effect of aging reflects primarily the cumulative increase of brain activity, especially the increase of mALFF in amygdala and mReHo in cingulate cortex, accompanied by the decrease of brain activity. The main effect of hypertension reflects primarily decreased brain activity in default modal network, accompanied by increased brain activity. The main effect of aging shows reduced brain FC as early as 24 weeks, and the main effect of hypertension shows higher brain FC in SHRs. The novel discovery is that 1 brain FC network increased linearly with age in SHRs, in addition to the linearly decreasing FC. Hypertension and aging independently contribute to spatiotemporal alterations in brain function in SHRs following ongoing progression and compensation. This study provides new insight into the dynamic characteristics of brain function.
Collapse
Affiliation(s)
- Yingying Yang
- Hebei Medical University Medical Imaging Specialty, Graduate School, , Shijiazhuang 050000 , China
- The First Hospital of Qinhuangdao Department of Imaging, , Qinhuangdao 066000 , China
| | - Qingfeng Zhu
- The Second Hospital of Hebei Medical University Department of Medical Imaging, , Shijiazhuang 050000 , China
| | - Lixin Wang
- The Second Hospital of Hebei Medical University Department of Medical Imaging, , Shijiazhuang 050000 , China
| | - Duo Gao
- The Second Hospital of Hebei Medical University Department of Medical Imaging, , Shijiazhuang 050000 , China
| | - Zhanqiu Wang
- The First Hospital of Qinhuangdao Department of Imaging, , Qinhuangdao 066000 , China
| | - Zuojun Geng
- The Second Hospital of Hebei Medical University Department of Medical Imaging, , Shijiazhuang 050000 , China
| |
Collapse
|
10
|
Grossmann K. Direct Oral Anticoagulants (DOACs) for Therapeutic Targeting of Thrombin, a Key Mediator of Cerebrovascular and Neuronal Dysfunction in Alzheimer's Disease. Biomedicines 2022; 10:1890. [PMID: 36009437 PMCID: PMC9405823 DOI: 10.3390/biomedicines10081890] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 07/29/2022] [Accepted: 07/30/2022] [Indexed: 11/16/2022] Open
Abstract
Although preclinical research and observer studies on patients with atrial fibrillation concluded that direct oral anticoagulants (DOACs) can protect against dementia like Alzheimer's disease (AD), clinical investigation towards therapeutical approval is still pending. DOACs target pathological thrombin, which is, like toxic tau and amyloid-ß proteins (Aß), an early hallmark of AD. Especially in hippocampal and neocortical areas, the release of parenchymal Aß into the blood induces thrombin and proinflammatory bradykinin synthesis by activating factor XII of the contact system. Thrombin promotes platelet aggregation and catalyzes conversion of fibrinogen to fibrin, leading to degradation-resistant, Aß-containing fibrin clots. Together with oligomeric Aß, these clots trigger vessel constriction and cerebral amyloid angiopathy (CAA) with vessel occlusion and hemorrhages, leading to vascular and blood-brain barrier (BBB) dysfunction. As consequences, brain blood flow, perfusion, and supply with oxygen (hypoxia) and nutrients decrease. In parenchymal tissue, hypoxia stimulates Aß synthesis, leading to Aß accumulation, which is further enhanced by BBB-impaired perivascular Aß clearance. Aß trigger neuronal damage and promote tau pathologies. BBB dysfunction enables thrombin and fibrin(ogen) to migrate into parenchymal tissue and to activate glial cells. Inflammation and continued Aß production are the results. Synapses and neurons die, and cognitive abilities are lost. DOACs block thrombin by inhibiting its activity (dabigatran) or production (FXa-inhibitors, e.g., apixaban, rivaroxaban). Therefore, DOAC use could preserve vascular integrity and brain perfusion and, thereby, could counteract vascular-driven neuronal and cognitive decline in AD. A conception for clinical investigation is presented, focused on DOAC treatment of patients with diagnosed AD in early-stage and low risk of major bleeding.
Collapse
Affiliation(s)
- Klaus Grossmann
- Center for Plant Molecular Biology (ZMBP), University of Tübingen, 72076 Tübingen, Germany
| |
Collapse
|
11
|
Neuroimaging of Mouse Models of Alzheimer’s Disease. Biomedicines 2022; 10:biomedicines10020305. [PMID: 35203515 PMCID: PMC8869427 DOI: 10.3390/biomedicines10020305] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/25/2022] [Accepted: 01/25/2022] [Indexed: 12/23/2022] Open
Abstract
Magnetic resonance imaging (MRI) and positron emission tomography (PET) have made great strides in the diagnosis and our understanding of Alzheimer’s Disease (AD). Despite the knowledge gained from human studies, mouse models have and continue to play an important role in deciphering the cellular and molecular evolution of AD. MRI and PET are now being increasingly used to investigate neuroimaging features in mouse models and provide the basis for rapid translation to the clinical setting. Here, we provide an overview of the human MRI and PET imaging landscape as a prelude to an in-depth review of preclinical imaging in mice. A broad range of mouse models recapitulate certain aspects of the human AD, but no single model simulates the human disease spectrum. We focused on the two of the most popular mouse models, the 3xTg-AD and the 5xFAD models, and we summarized all known published MRI and PET imaging data, including contrasting findings. The goal of this review is to provide the reader with broad framework to guide future studies in existing and future mouse models of AD. We also highlight aspects of MRI and PET imaging that could be improved to increase rigor and reproducibility in future imaging studies.
Collapse
|
12
|
Ali M, Falkenhain K, Njiru BN, Murtaza-Ali M, Ruiz-Uribe NE, Haft-Javaherian M, Catchers S, Nishimura N, Schaffer CB, Bracko O. VEGF signalling causes stalls in brain capillaries and reduces cerebral blood flow in Alzheimer's mice. Brain 2022; 145:1449-1463. [PMID: 35048960 PMCID: PMC9150081 DOI: 10.1093/brain/awab387] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 07/09/2021] [Accepted: 09/22/2021] [Indexed: 11/16/2022] Open
Abstract
Increased incidence of stalled capillary blood flow caused by adhesion of
leucocytes to the brain microvascular endothelium leads to a 17%
reduction of cerebral blood flow and exacerbates short-term memory loss in
multiple mouse models of Alzheimer’s disease. Here, we report that vascular endothelial growth factor (VEGF) signalling at the
luminal side of the brain microvasculature plays an integral role in the
capillary stalling phenomenon of the APP/PS1 mouse model. Administration of the anti-mouse VEGF-A164 antibody, an isoform that inhibits
blood–brain barrier hyperpermeability, reduced the number of stalled
capillaries within an hour of injection, leading to an immediate increase in
average capillary blood flow but not capillary diameter. VEGF-A inhibition also
reduced the overall endothelial nitric oxide synthase protein concentrations,
increased occludin levels and decreased the penetration of circulating Evans
Blue dye across the blood–brain barrier into the brain parenchyma,
suggesting increased blood–brain barrier integrity. Capillaries prone to
neutrophil adhesion after anti-VEGF-A treatment also had lower occludin
concentrations than flowing capillaries. Taken together, our findings demonstrate that VEGF-A signalling in APP/PS1 mice
contributes to aberrant endothelial nitric oxide synthase /occludin-associated
blood–brain barrier permeability, increases the incidence of capillary
stalls, and leads to reductions in cerebral blood flow. Reducing leucocyte
adhesion by inhibiting luminal VEGF signalling may provide a novel and
well-tolerated strategy for improving brain microvascular blood flow in
Alzheimer’s disease patients.
Collapse
Affiliation(s)
- Muhammad Ali
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA, 148532
| | - Kaja Falkenhain
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA, 148532
| | - Brendah N Njiru
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA, 148532
| | - Muhammad Murtaza-Ali
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA, 148532
| | - Nancy E Ruiz-Uribe
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA, 148532
| | | | | | - Nozomi Nishimura
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA, 148532
| | - Chris B Schaffer
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA, 148532
| | - Oliver Bracko
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA, 148532
| |
Collapse
|
13
|
Wang C, Lin G, Shen Z, Wang R. Angiopep-2 as an Exogenous Chemical Exchange Saturation Transfer Contrast Agent in Diagnosis of Alzheimer's Disease. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:7480519. [PMID: 35422975 PMCID: PMC9005290 DOI: 10.1155/2022/7480519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 02/22/2022] [Accepted: 03/11/2022] [Indexed: 02/05/2023]
Abstract
BACKGROUND Chemical exchange saturation transfer (CEST) is a novel imaging modality in clinical practice and scientific research. Angiopep-2 is an artificial peptide that can penetrate blood-brain barrier. The aim of this study was to explore the feasibility of Angiopep-2 serving as an exogenous CEST contrast. METHODS Phantoms of Angiopep-2 with different concentrations were prepared and then scanned using the 7.0T small animal MRI scanner. Different parameters including saturation powers and saturation duration were used to achieve the optimal CEST effect, and the optimal parameters were finally selected based on Z-spectra, asymmetric spectra, and phantom CEST imaging. CEST scanning of dimethyl sulfoxide (DMSO), the substance helping Angiopep-2 to be dissolved in water, was performed to exclude its contribution for the CEST effect. RESULTS A broad dip was observed from 2.5 to 3.5 ppm in the Z-spectra of Angiopep-2 phantoms. The most robust CEST was generated at 3.2 ppm when using formula (M -3.2ppm - M +3.2ppm)/M -3.2ppm. The CEST effect of Angiopep-2 was concentration dependent; the effect increased as the concentration increased. In addition, the CEST effect was more obvious as the saturation power increased and peaked at 5.5 µT, and the CEST effect increased as the saturation duration increased. DMSO showed nearly 0% of the CEST effect at 3.2 ppm. CONCLUSIONS Our results demonstrate that Angiopep-2 can act as an excellent exogenous CEST contrast. As it can penetrate blood-brain barrier and bind amyloid-β protein, amyloid-β targeting CEST, with Angiopep-2 as an exogenous contrast agent, can be potentially used as a novel imaging modality for early diagnosis of Alzheimer's disease. Collectively, Angiopep-2 may play a critical role in early diagnosis of Alzheimer's disease.
Collapse
Affiliation(s)
- Chengguang Wang
- Department of Neurosurgery, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, Henan, China
| | - Guisen Lin
- Department of Medical Imaging, 2nd Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515000, China
| | - Zhiwei Shen
- Philips Healthcare, Tianzhe Road 16, Chaoyang, Beijing 100600, China
| | - Runrun Wang
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, Henan, China
| |
Collapse
|
14
|
Ali M, Bracko O. VEGF Paradoxically Reduces Cerebral Blood Flow in Alzheimer’s Disease Mice. Neurosci Insights 2022; 17:26331055221109254. [PMID: 35873789 PMCID: PMC9298729 DOI: 10.1177/26331055221109254] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 12/19/2022] Open
Abstract
Vascular dysfunction plays a critical role in the development of Alzheimer’s disease. Cerebral blood flow reductions of 10% to 25% present early in disease pathogenesis. Vascular Endothelial Growth Factor-A (VEGF-A) drives angiogenesis, which typically addresses blood flow reductions and global hypoxia. However, recent evidence suggests aberrant VEGF-A signaling in Alzheimer’s disease may undermine its physiological angiogenic function. Instead of improving cerebral blood flow, VEGF-A contributes to brain capillary stalls and blood flow reductions, likely accelerating cognitive decline. In this commentary, we explore the evidence for pathological VEGF signaling in Alzheimer’s disease, and discuss its implications for disease therapy.
Collapse
Affiliation(s)
- Muhammad Ali
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Oliver Bracko
- Department of Biology, University of Miami, Coral Gables, FL, USA
| |
Collapse
|
15
|
Ni R. Magnetic Resonance Imaging in Animal Models of Alzheimer's Disease Amyloidosis. Int J Mol Sci 2021; 22:12768. [PMID: 34884573 PMCID: PMC8657987 DOI: 10.3390/ijms222312768] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/18/2021] [Accepted: 11/23/2021] [Indexed: 02/07/2023] Open
Abstract
Amyloid-beta (Aβ) plays an important role in the pathogenesis of Alzheimer's disease. Aberrant Aβ accumulation induces neuroinflammation, cerebrovascular alterations, and synaptic deficits, leading to cognitive impairment. Animal models recapitulating the Aβ pathology, such as transgenic, knock-in mouse and rat models, have facilitated the understanding of disease mechanisms and the development of therapeutics targeting Aβ. There is a rapid advance in high-field MRI in small animals. Versatile high-field magnetic resonance imaging (MRI) sequences, such as diffusion tensor imaging, arterial spin labeling, resting-state functional MRI, anatomical MRI, and MR spectroscopy, as well as contrast agents, have been developed for preclinical imaging in animal models. These tools have enabled high-resolution in vivo structural, functional, and molecular readouts with a whole-brain field of view. MRI has been used to visualize non-invasively the Aβ deposits, synaptic deficits, regional brain atrophy, impairment in white matter integrity, functional connectivity, and cerebrovascular and glymphatic system in animal models of Alzheimer's disease amyloidosis. Many of the readouts are translational toward clinical MRI applications in patients with Alzheimer's disease. In this review, we summarize the recent advances in MRI for visualizing the pathophysiology in amyloidosis animal models. We discuss the outstanding challenges in brain imaging using MRI in small animals and propose future outlook in visualizing Aβ-related alterations in the brains of animal models.
Collapse
Affiliation(s)
- Ruiqing Ni
- Institute for Biomedical Engineering, ETH Zurich & University of Zurich, 8093 Zurich, Switzerland;
- Institute for Regenerative Medicine, University of Zurich, 8952 Zurich, Switzerland
| |
Collapse
|
16
|
Rivera DS, Lindsay CB, Oliva CA, Bozinovic F, Inestrosa NC. A Multivariate Assessment of Age-Related Cognitive Impairment in Octodon degus. Front Integr Neurosci 2021; 15:719076. [PMID: 34526882 PMCID: PMC8437396 DOI: 10.3389/fnint.2021.719076] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 07/23/2021] [Indexed: 01/27/2023] Open
Abstract
Aging is a progressive functional decline characterized by a gradual deterioration in physiological function and behavior. The most important age-related change in cognitive function is decline in cognitive performance (i.e., the processing or transformation of information to make decisions that includes speed of processing, working memory, and learning). The purpose of this study is to outline the changes in age-related cognitive performance (i.e., short-term recognition memory and long-term learning and memory) in long-lived Octodon degus. The strong similarity between degus and humans in social, metabolic, biochemical, and cognitive aspects makes it a unique animal model for exploring the mechanisms underlying the behavioral and cognitive deficits related to natural aging. In this study, we examined young adult female degus (12- and 24-months-old) and aged female degus (38-, 56-, and 75-months-old) that were exposed to a battery of cognitive-behavioral tests. Multivariate analyses of data from the Social Interaction test or Novel Object/Local Recognition (to measure short-term recognition memory), and the Barnes maze test (to measure long-term learning and memory) revealed a consistent pattern. Young animals formed a separate group of aged degus for both short- and long-term memories. The association between the first component of the principal component analysis (PCA) from short-term memory with the first component of the PCA from long-term memory showed a significant negative correlation. This suggests age-dependent differences in both memories, with the aged degus having higher values of long-term memory ability but poor short-term recognition memory, whereas in the young degus an opposite pattern was found. Approximately 5% of the young and 80% of the aged degus showed an impaired short-term recognition memory; whereas for long-term memory about 32% of the young degus and 57% of the aged degus showed decreased performance on the Barnes maze test. Throughout this study, we outlined age-dependent cognitive performance decline during natural aging in degus. Moreover, we also demonstrated that the use of a multivariate approach let us explore and visualize complex behavioral variables, and identified specific behavioral patterns that allowed us to make powerful conclusions that will facilitate further the study on the biology of aging. In addition, this study could help predict the onset of the aging process based on behavioral performance.
Collapse
Affiliation(s)
- Daniela S Rivera
- GEMA Center for Genomics, Ecology and Environment, Facultad de Estudios Interdisciplinarios, Universidad Mayor, Santiago, Chile
| | - Carolina B Lindsay
- Center of Aging and Regeneration UC (CARE-UC), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carolina A Oliva
- Center of Aging and Regeneration UC (CARE-UC), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Francisco Bozinovic
- Center for Applied Ecology and Sustainability (CAPES), Departamento de Ecología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nibaldo C Inestrosa
- Center of Aging and Regeneration UC (CARE-UC), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
| |
Collapse
|
17
|
Grossmann K. Alzheimer's Disease-Rationales for Potential Treatment with the Thrombin Inhibitor Dabigatran. Int J Mol Sci 2021; 22:ijms22094805. [PMID: 33946588 PMCID: PMC8125318 DOI: 10.3390/ijms22094805] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 04/28/2021] [Accepted: 04/28/2021] [Indexed: 12/16/2022] Open
Abstract
Alzheimer's disease (AD) is caused by neurodegenerative, but also vascular and hemostatic changes in the brain. The oral thrombin inhibitor dabigatran, which has been used for over a decade in preventing thromboembolism and has a well-known pharmacokinetic, safety and antidote profile, can be an option to treat vascular dysfunction in early AD, a condition known as cerebral amyloid angiopathy (CAA). Recent results have revealed that amyloid-β proteins (Aβ), thrombin and fibrin play a crucial role in triggering vascular and parenchymal brain abnormalities in CAA. Dabigatran blocks soluble thrombin, thrombin-mediated formation of fibrin and Aβ-containing fibrin clots. These clots are deposited in brain parenchyma and blood vessels in areas of CAA. Fibrin-Aβ deposition causes microvascular constriction, occlusion and hemorrhage, leading to vascular and blood-brain barrier dysfunction. As a result, blood flow, perfusion and oxygen and nutrient supply are chronically reduced, mainly in hippocampal and neocortical brain areas. Dabigatran has the potential to preserve perfusion and oxygen delivery to the brain, and to prevent parenchymal Aβ-, thrombin- and fibrin-triggered inflammatory and neurodegenerative processes, leading to synapse and neuron death, and cognitive decline. Beneficial effects of dabigatran on CAA and AD have recently been shown in preclinical studies and in retrospective observer studies on patients. Therefore, clinical studies are warranted, in order to possibly expand dabigatran approval for repositioning for AD treatment.
Collapse
Affiliation(s)
- Klaus Grossmann
- Center for Plant Molecular Biology (ZMBP), University of Tübingen, 72076 Tübingen, Germany
| |
Collapse
|
18
|
Sex-Dependent End-of-Life Mental and Vascular Scenarios for Compensatory Mechanisms in Mice with Normal and AD-Neurodegenerative Aging. Biomedicines 2021; 9:biomedicines9020111. [PMID: 33498895 PMCID: PMC7911097 DOI: 10.3390/biomedicines9020111] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 01/17/2021] [Accepted: 01/20/2021] [Indexed: 02/07/2023] Open
Abstract
Life expectancy decreases with aging, with cardiovascular, mental health, and neurodegenerative disorders strongly contributing to the total disability-adjusted life years. Interestingly, the morbidity/mortality paradox points to females having a worse healthy life expectancy. Since bidirectional interactions between cardiovascular and Alzheimer’s diseases (AD) have been reported, the study of this emerging field is promising. In the present work, we further explored the cardiovascular–brain interactions in mice survivors of two cohorts of non-transgenic and 3xTg-AD mice, including both sexes, to investigate the frailty/survival through their life span. Survival, monitored from birth, showed exceptionally worse mortality rates in females than males, independently of the genotype. This mortality selection provided a “survivors” cohort that could unveil brain–cardiovascular interaction mechanisms relevant for normal and neurodegenerative aging processes restricted to long-lived animals. The results show sex-dependent distinct physical (worse in 3xTg-AD males), neuropsychiatric-like and cognitive phenotypes (worse in 3xTg-AD females), and hypothalamic–pituitary–adrenal (HPA) axis activation (higher in females), with higher cerebral blood flow and improved cardiovascular phenotype in 3xTg-AD female mice survivors. The present study provides an experimental scenario to study the suggested potential compensatory hemodynamic mechanisms in end-of-life dementia, which is sex-dependent and can be a target for pharmacological and non-pharmacological interventions.
Collapse
|
19
|
Stiebing C, Jahn IJ, Schmitt M, Keijzer N, Kleemann R, Kiliaan AJ, Drexler W, Leitgeb RA, Popp J. Biochemical Characterization of Mouse Retina of an Alzheimer's Disease Model by Raman Spectroscopy. ACS Chem Neurosci 2020; 11:3301-3308. [PMID: 32991138 PMCID: PMC7581290 DOI: 10.1021/acschemneuro.0c00420] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
![]()
The presence of biomarkers characteristic
for Alzheimer’s
disease in the retina is a controversial topic. Raman spectroscopy
offers information on the biochemical composition of tissues. Thus,
it could give valuable insight into the diagnostic value of retinal
analysis. Within the present study, retinas of a double transgenic
mouse model, that expresses a chimeric mouse/human amyloid precursor
protein and a mutant form of human presenilin 1, and corresponding
control group were subjected to ex vivo Raman imaging.
The Raman data recorded on cross sections of whole eyes highlight
the layered structure of the retina in a label-free manner. Based
on the Raman information obtained from en face mounted
retina samples, a discrimination between healthy and Alzheimer’s
disease retinal tissue can be done with an accuracy of 85.9%. For
this a partial least squares-linear discriminant analysis was applied.
Therefore, although no macromolecular changes in form of, i.e., amyloid beta plaques, can be noticed based on Raman
spectroscopy, subtle biochemical changes happening in the retina could
lead to Alzheimer’s disease identification.
Collapse
Affiliation(s)
- Clara Stiebing
- Leibniz Institute of Photonic Technology (Leibniz-IPHT), a member of the Leibniz Research Alliance Leibniz Health Technology, Albert-Einstein-Straße 9, 07745 Jena, Germany
| | - Izabella J. Jahn
- Leibniz Institute of Photonic Technology (Leibniz-IPHT), a member of the Leibniz Research Alliance Leibniz Health Technology, Albert-Einstein-Straße 9, 07745 Jena, Germany
| | - Michael Schmitt
- Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich Schiller University Jena, Helmholtzweg 4, 07743 Jena, Germany
| | - Nanda Keijzer
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), Zernikedreef 9, 2333 CK Leiden, The Netherlands
| | - Robert Kleemann
- Department of Metabolic Health Research, Netherlands Organisation for Applied Scientific Research (TNO), Zernikedreef 9, 2333 CK Leiden, The Netherlands
| | - Amanda J. Kiliaan
- Department of Anatomy Donders Institute for Brain, Cognition, and Behavior Preclinical Imaging Center, Radboud University Medical Center, Geert Grooteplein 21N, 6525 EZ Nijmegen, The Netherlands
| | - Wolfgang Drexler
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Waehringerguertel 18-20, 1090 Vienna, Austria
| | - Rainer A. Leitgeb
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Waehringerguertel 18-20, 1090 Vienna, Austria
| | - Jürgen Popp
- Leibniz Institute of Photonic Technology (Leibniz-IPHT), a member of the Leibniz Research Alliance Leibniz Health Technology, Albert-Einstein-Straße 9, 07745 Jena, Germany
- Institute of Physical Chemistry and Abbe Center of Photonics, Friedrich Schiller University Jena, Helmholtzweg 4, 07743 Jena, Germany
| |
Collapse
|
20
|
Huang Y, Zheng H, Tan K, Sun X, Ye J, Zhang Y. Circulating metabolomics profiling reveals novel pathways associated with cognitive decline in patients with hypertension. Ther Adv Neurol Disord 2020; 13:1756286420947973. [PMID: 32952614 PMCID: PMC7476355 DOI: 10.1177/1756286420947973] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 07/16/2020] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Hypertension is a significant risk factor for cardiovascular disease, and it is associated with dementia, including Alzheimer's disease (AD). Although it may be correlated with AD in terms of symptoms, the link between hypertension and AD pathological biomarkers, and the potential underlying mechanism of hypertension with cognitive decline, are still not well understood. METHODS The Mini-Mental State Examination (MMSE) scores were used to evaluate cognitive function. Enzyme-linked immunosorbent assays were used to examine plasma amyloid-beta (Aβ)40, Aβ42, and tau concentration in hypertensive patients. Metabolomics and metagenomics were performed to identify the significantly changed circulating metabolites and microbiota between healthy individuals and hypertensive patients. Pearson's correlation was used to examine the association between cognitive indicators and differential metabolites. RESULTS We found significantly decreased MMSE scores, elevated plasma Aβ40, and decreased Aβ42/Aβ40 ratio in hypertensive patients, which are critically associated with AD pathology. Based on metabolomics, we found that significantly altered metabolites in the plasma of hypertensive patients were enriched in the benzoate degradation and phenylpropanoid biosynthesis pathways, and they were also correlated with changes in MMSE scores and Aβ42/Aβ40 ratio. In addition, metabolomics signaling pathway analysis suggested that microbial metabolism was altered in hypertensive patients. We also identified altered blood microbiota in hypertensive patients compared with the controls. CONCLUSIONS Our study provides a novel metabolic and microbial mechanism, which may underlie the cognitive impairment in hypertensive patients.
Collapse
Affiliation(s)
- Yuli Huang
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde Foshan), Foshan, China
| | - Haoxiao Zheng
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde Foshan), Foshan, China
| | - Kuan Tan
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde Foshan), Foshan, China
| | - Xiangdong Sun
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| | - Jinshao Ye
- Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou, 510632, China
| | - Yunlong Zhang
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Panyu District, Xinzao, Guangzhou, 510260, China
| |
Collapse
|
21
|
Falkenhain K, Ruiz-Uribe NE, Haft-Javaherian M, Ali M, Michelucci PE, Schaffer CB, Bracko O. A pilot study investigating the effects of voluntary exercise on capillary stalling and cerebral blood flow in the APP/PS1 mouse model of Alzheimer's disease. PLoS One 2020; 15:e0235691. [PMID: 32857763 PMCID: PMC7455035 DOI: 10.1371/journal.pone.0235691] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 06/19/2020] [Indexed: 11/18/2022] Open
Abstract
Exercise exerts a beneficial effect on the major pathological and clinical symptoms associated with Alzheimer’s disease in humans and mouse models of the disease. While numerous mechanisms for such benefits from exercise have been proposed, a clear understanding of the causal links remains elusive. Recent studies also suggest that cerebral blood flow in the brain of both Alzheimer’s patients and mouse models of the disease is decreased and that the cognitive symptoms can be improved when blood flow is restored. We therefore hypothesized that the mitigating effect of exercise on the development and progression of Alzheimer’s disease may be mediated through an increase in the otherwise reduced brain blood flow. To test this idea, we performed a pilot study to examine the impact of three months of voluntary wheel running in a small cohort of ~1-year-old APP/PS1 mice on short-term memory function, brain inflammation, amyloid deposition, and baseline cerebral blood flow. Our findings that exercise led to a trend toward improved spatial short-term memory, reduced brain inflammation, markedly increased neurogenesis in the dentate gyrus, and a reduction in hippocampal amyloid-beta deposits are consistent with other reports on the impact of exercise on the progression of Alzheimer’s related symptoms in mouse models. Notably, we did not observe any impact of wheel running on overall baseline blood flow nor on the incidence of non-flowing capillaries, a mechanism we recently identified as one contributing factor to cerebral blood flow deficits in mouse models of Alzheimer’s disease. Overall, our findings add to the emerging picture of differential effects of exercise on cognition and blood flow in Alzheimer’s disease pathology by showing that capillary stalling is not decreased following exercise.
Collapse
Affiliation(s)
- Kaja Falkenhain
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States of America
- Institute of Cognitive Science, Osnabrück University, Osnabrück, Germany
| | - Nancy E. Ruiz-Uribe
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States of America
| | - Mohammad Haft-Javaherian
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States of America
| | - Muhammad Ali
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States of America
| | | | | | - Chris B. Schaffer
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States of America
| | - Oliver Bracko
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States of America
- * E-mail:
| |
Collapse
|
22
|
Liu F, Tian N, Zhang HQ, Li SH, Zhou QZ, Yang Y, Zheng J, Wang JZ. GSK-3β activation accelerates early-stage consumption of Hippocampal Neurogenesis in senescent mice. Theranostics 2020; 10:9674-9685. [PMID: 32863953 PMCID: PMC7449917 DOI: 10.7150/thno.43829] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 07/09/2020] [Indexed: 12/27/2022] Open
Abstract
Adult hippocampal neurogenesis (AHN) deficits contribute to the progression of cognitive impairments during accelerated senescence, with the mechanistic causes poorly understood. Glycogen synthase kinase-3β (GSK-3β) is a critical regulator in prenatal neurodevelopment. The present study aims to study whether and how GSK-3β regulates AHN during the accelerated senescence. Methods: AHN and AHN-dependent cognition and GSK-3β were evaluated in 3- and 6-month senescence-accelerated mice prone 8 (SAM-P8) and senescence resistant 1 (SAM-R1) mice, respectively. GSK-3β was selectively overexpressed in wild-type mice using adeno-associated virus, or knocked-out by crossbreeding with GSK-3β floxed mice in the neural stem cells (NSCs) of Nestin-Cre mice, or pharmacologically inhibited with SB216763 in SAM-P8 mice. AHN was evaluated by BrdU-, DCX-staining and retrovirus-labeling. Results: AHN transiently increased at 3-month, but dramatically dropped at 6-month of age in SAM-P8 mice with a simultaneous activation of GSK-3β at 3-month. Selective overexpression of GSK-3β in hippocampal NSCs of wildtype mice induced long-term AHN deficits due to an accelerated depletion of NSC pool, although it transiently increased the proliferation and survival of the newborn neurons. Pharmacologically inhibiting GSK-3β by SB216763 efficiently preserved AHN and improved contextual memory in 6-month SAM-P8 mice, while conditional knock-out of GSK-3β in NSCs impaired AHN. Conclusion: Early-stage activation of GSK-3β in NSCs impairs AHN by accelerating the depletion of NSC pool, and pharmacological inhibition of GSK-3β is efficient to preserve AHN during the accelerated aging. These results reveal novel mechanisms underlying the AHN impairments during accelerated senescence and provide new targets for pro-neurogenic therapies for related diseases.
Collapse
Affiliation(s)
- Fei Liu
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Department of Human Anatomy, School of Basic Medicine, Binzhou Medical University, Yantai 264003, China
| | - Na Tian
- Department of Histology and Embryology, Key Laboratory of Ministry of Education of China for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hua-Qiu Zhang
- Key Laboratory of Ministry of Education for Neurological Disorders, Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shi-Hong Li
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qiu-Zhi Zhou
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ying Yang
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jie Zheng
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jian-Zhi Wang
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Key Laboratory of Ministry of Education for Neurological Disorders, Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226000, China
| |
Collapse
|
23
|
Kerkhofs D, van Hagen BT, Milanova IV, Schell KJ, van Essen H, Wijnands E, Goossens P, Blankesteijn WM, Unger T, Prickaerts J, Biessen EA, van Oostenbrugge RJ, Foulquier S. Pharmacological depletion of microglia and perivascular macrophages prevents Vascular Cognitive Impairment in Ang II-induced hypertension. Am J Cancer Res 2020; 10:9512-9527. [PMID: 32863942 PMCID: PMC7449902 DOI: 10.7150/thno.44394] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 06/12/2020] [Indexed: 12/19/2022] Open
Abstract
Rationale: Hypertension is a major risk factor for cerebral small vessel disease, the most prevalent cause of vascular cognitive impairment. As we have shown, hypertension induced by a prolonged Angiotensin II infusion is associated with increased permeability of the blood-brain barrier (BBB), chronic activation of microglia and myelin loss. In this study we therefore aim to determine the contribution of microglia to hypertension-induced cognitive impairment in an experimental hypertension model by a pharmacological depletion approach. Methods: For this study, adult Cx3Cr1gfp/wtxThy1yfp/0 reporter mice were infused for 12 weeks with Angiotensin II or saline and subgroups were treated with PLX5622, a highly selective CSF1R tyrosine kinase inhibitor. Systolic blood pressure (SBP) was measured via tail-cuff. Short- and long-term spatial memory was assessed during an Object Location task and a Morris Water Maze task (MWM). Microglia depletion efficacy was assessed by flow cytometry and immunohistochemistry. BBB leakages, microglia phenotype and myelin integrity were assessed by immunohistochemistry. Results: SBP, heart weight and carotid pulsatility were increased by Ang II and were not affected by PLX5622. Short-term memory was significantly impaired in Ang II hypertensive mice, and partly prevented in Ang II mice treated with PLX5622. Histological and flow cytometry analysis revealed almost complete ablation of microglia and a 60% depletion of brain resident perivascular macrophages upon CSF1R inhibition. Number and size of BBB leakages were increased in Ang II hypertensive mice, but not altered by PLX5622 treatment. Microglia acquired a pro-inflammatory phenotype at the site of BBB leakages in both Saline and Ang II mice and were successfully depleted by PLX5622. There was however no significant change in myelin integrity at the site of leakages. Conclusion: Our results show that depletion of microglia and PVMs, by CSF1R inhibition prevents short-term memory impairment in Ang II induced hypertensive mice. We suggest this beneficial effect is mediated by the major decrease of pro-inflammatory microglia within BBB leakages. This novel finding supports the critical role of brain immune cells in the pathogenesis of hypertension-related cognitive impairment. An adequate modulation of microglia /PVM density and phenotype may constitute a relevant approach to prevent and/or limit the progression of vascular cognitive impairment.
Collapse
|
24
|
Bracko O, Njiru BN, Swallow M, Ali M, Haft-Javaherian M, Schaffer CB. Increasing cerebral blood flow improves cognition into late stages in Alzheimer's disease mice. J Cereb Blood Flow Metab 2020; 40:1441-1452. [PMID: 31495298 PMCID: PMC7308509 DOI: 10.1177/0271678x19873658] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease is associated with a 20-30% reduction in cerebral blood flow. In the APP/PS1 mouse model of Alzheimer's disease, inhibiting neutrophil adhesion using an antibody against the neutrophil specific protein Ly6G was recently shown to drive rapid improvements in cerebral blood flow that was accompanied by an improvement in performance on short-term memory tasks. Here, in a longitudinal aging study, we assessed how far into disease development a single injection of anti-Ly6G treatment can acutely improve short-term memory function. We found that APP/PS1 mice as old as 15-16 months had improved performance on the object replacement and Y-maze tests of spatial and working short-term memory, measured at one day after anti-Ly6G treatment. APP/PS1 mice at 17-18 months of age or older did not show acute improvements in cognitive performance, although we did find that capillary stalls were still reduced and cerebral blood flow was still increased by 17% in 21-22-months-old APP/PS1 mice given anti-Ly6G antibody. These data add to the growing body of evidence suggesting that cerebral blood flow reductions are an important contributing factor to the cognitive dysfunction associated with neurodegenerative disease. Thus, interfering with neutrophil adhesion could be a new therapeutic approach for Alzheimer's disease.
Collapse
Affiliation(s)
- Oliver Bracko
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Brendah N Njiru
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Madisen Swallow
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Muhammad Ali
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Mohammad Haft-Javaherian
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Chris B Schaffer
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| |
Collapse
|
25
|
Tengeler AC, Gart E, Wiesmann M, Arnoldussen IAC, van Duyvenvoorde W, Hoogstad M, Dederen PJ, Verweij V, Geenen B, Kozicz T, Kleemann R, Morrison MC, Kiliaan AJ. Propionic acid and not caproic acid, attenuates nonalcoholic steatohepatitis and improves (cerebro) vascular functions in obese Ldlr -/- .Leiden mice. FASEB J 2020; 34:9575-9593. [PMID: 32472598 DOI: 10.1096/fj.202000455r] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 05/07/2020] [Accepted: 05/11/2020] [Indexed: 02/07/2023]
Abstract
The obesity epidemic increases the interest to elucidate impact of short-chain fatty acids on metabolism, obesity, and the brain. We investigated the effects of propionic acid (PA) and caproic acid (CA) on metabolic risk factors, liver and adipose tissue pathology, brain function, structure (by MRI), and gene expression, during obesity development in Ldlr-/- .Leiden mice. Ldlr-/- .Leiden mice received 16 weeks either a high-fat diet (HFD) to induce obesity, or chow as reference group. Next, obese HFD-fed mice were treated 12 weeks with (a) HFD + CA (CA), (b) HFD + PA (PA), or (c) a HFD-control group. PA reduced the body weight and systolic blood pressure, lowered fasting insulin levels, and reduced HFD-induced liver macrovesicular steatosis, hypertrophy, inflammation, and collagen content. PA increased the amount of glucose transporter type 1-positive cerebral blood vessels, reverted cerebral vasoreactivity, and HFD-induced effects in microstructural gray and white matter integrity of optic tract, and somatosensory and visual cortex. PA and CA also reverted HFD-induced effects in functional connectivity between visual and auditory cortex. However, PA mice were more anxious in open field, and showed reduced activity of synaptogenesis and glutamate regulators in hippocampus. Therefore, PA treatment should be used with caution even though positive metabolic, (cerebro) vascular, and brain structural and functional effects were observed.
Collapse
Affiliation(s)
- Anouk C Tengeler
- Department of Anatomy, Donders Institute for Brain, Cognition and Behavior, Preclinical Imaging Centre, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Eveline Gart
- Department of Metabolic Health Research, The Netherlands Organisation for Applied Scientific Research (TNO), Leiden, the Netherlands.,Human and Animal Physiology, Wageningen University, Wageningen, the Netherlands
| | - Maximilian Wiesmann
- Department of Anatomy, Donders Institute for Brain, Cognition and Behavior, Preclinical Imaging Centre, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Ilse A C Arnoldussen
- Department of Anatomy, Donders Institute for Brain, Cognition and Behavior, Preclinical Imaging Centre, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Wim van Duyvenvoorde
- Department of Metabolic Health Research, The Netherlands Organisation for Applied Scientific Research (TNO), Leiden, the Netherlands
| | - Marloes Hoogstad
- Department of Anatomy, Donders Institute for Brain, Cognition and Behavior, Preclinical Imaging Centre, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Pieter J Dederen
- Department of Anatomy, Donders Institute for Brain, Cognition and Behavior, Preclinical Imaging Centre, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Vivienne Verweij
- Department of Anatomy, Donders Institute for Brain, Cognition and Behavior, Preclinical Imaging Centre, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Bram Geenen
- Department of Anatomy, Donders Institute for Brain, Cognition and Behavior, Preclinical Imaging Centre, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Tamas Kozicz
- Department of Anatomy, Donders Institute for Brain, Cognition and Behavior, Preclinical Imaging Centre, Radboud University Medical Center, Nijmegen, the Netherlands.,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Robert Kleemann
- Department of Anatomy, Donders Institute for Brain, Cognition and Behavior, Preclinical Imaging Centre, Radboud University Medical Center, Nijmegen, the Netherlands.,Department of Vascular Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Martine C Morrison
- Department of Metabolic Health Research, The Netherlands Organisation for Applied Scientific Research (TNO), Leiden, the Netherlands.,Human and Animal Physiology, Wageningen University, Wageningen, the Netherlands
| | - Amanda J Kiliaan
- Department of Anatomy, Donders Institute for Brain, Cognition and Behavior, Preclinical Imaging Centre, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
26
|
Han B, Jiang W, Liu H, Wang J, Zheng K, Cui P, Feng Y, Dang C, Bu Y, Wang QM, Ju Z, Hao J. Upregulation of neuronal PGC-1α ameliorates cognitive impairment induced by chronic cerebral hypoperfusion. Am J Cancer Res 2020; 10:2832-2848. [PMID: 32194838 PMCID: PMC7052889 DOI: 10.7150/thno.37119] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 09/17/2019] [Indexed: 12/18/2022] Open
Abstract
Rationale: Mitochondrial dysfunction and oxidative stress occur in vascular dementia (VaD), but the specific molecular mechanism regulating these events remains unclear. Peroxisome proliferator-activated receptor-γ co-activator-1α (PGC-1α) is a master regulator for mitochondrial function. This study aims to investigate whether PGC-1α is involved in the pathophysiology of VaD. Methods: We firstly generated PGC-1α f/f Eno2-Cre mice to induce neuron-specific overexpression of PGC-1α by crossbreeding PGC-1α f/f mice with Eno2-cre mice. Then, the mice were subjected to bilateral common carotid artery stenosis to induce chronic cerebral hypoperfusion. Neurological function and hippocampal PGC-1α expression was evaluated. Next, RNA-Seq analysis and Seahorse assay were performed on the hippocampal neurons. In addition, mitochondrial antioxidants, uncoupling proteins, ROS production and the activation of glial cells were also measured. Results: Our results showed that hippocampal PGC-1α expression is down-regulated in the mouse VaD model induced by chronic cerebral hypoperfusion. In contrast, neuronal PGC-1α overexpression significantly ameliorated cognitive deficits. RNA-Seq analysis indicated that PGC-1α improved energy metabolism of neurons under hypoxic condition, and Seahorse assay confirmed that PGC-1α increases the metabolic activity of neurons. Further study demonstrated that PGC-1α boosted the expressions of mitochondrial antioxidants and uncoupling proteins (UCPs), including SOD2, Prx3, GPx1, UCP2, UCP4 and UCP5, which in turn reduced reactive oxygen species (ROS) production. Moreover, the activation of microglia and astrocytes was also found to decrease in the hippocampus. All of these changes greatly contributed to protect hippocampal neurons against ischemic insults. Conclusions: PGC-1α could suppress the excessive ROS and neuroinflammation in the hippocampus, opening up a potential therapeutic target for cognitive impairment.
Collapse
|
27
|
Solis E, Hascup KN, Hascup ER. Alzheimer's Disease: The Link Between Amyloid-β and Neurovascular Dysfunction. J Alzheimers Dis 2020; 76:1179-1198. [PMID: 32597813 PMCID: PMC7483596 DOI: 10.3233/jad-200473] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
While prevailing evidence supports that the amyloid cascade hypothesis is a key component of Alzheimer's disease (AD) pathology, many recent studies indicate that the vascular system is also a major contributor to disease progression. Vascular dysfunction and reduced cerebral blood flow (CBF) occur prior to the accumulation and aggregation of amyloid-β (Aβ) plaques and hyperphosphorylated tau tangles. Although research has predominantly focused on the cellular processes involved with Aβ-mediated neurodegeneration, effects of Aβ on CBF and neurovascular coupling are becoming more evident. This review will describe AD vascular disturbances as they relate to Aβ, including chronic cerebral hypoperfusion, hypertension, altered neurovascular coupling, and deterioration of the blood-brain barrier. In addition, we will describe recent findings about the relationship between these vascular defects and Aβ accumulation with emphasis on in vivo studies utilizing rodent AD models.
Collapse
Affiliation(s)
- Ernesto Solis
- Department of Neurology, Neuroscience Institute, Center for Alzheimer’s Disease and Related Disorders, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Kevin N. Hascup
- Department of Neurology, Neuroscience Institute, Center for Alzheimer’s Disease and Related Disorders, Southern Illinois University School of Medicine, Springfield, IL, USA
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Erin R. Hascup
- Department of Neurology, Neuroscience Institute, Center for Alzheimer’s Disease and Related Disorders, Southern Illinois University School of Medicine, Springfield, IL, USA
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA
| |
Collapse
|
28
|
Abstract
Alzheimer's disease (AD) is a multifactorial syndrome with a plethora of progressive, degenerative changes in the brain parenchyma, but also in the cerebrovascular and hemostatic system. A therapeutic approach for AD is reviewed, which is focused on the role of amyloid-β protein (Aβ) and fibrin in triggering intra-brain vascular dysfunction and connected, cognitive decline. It is proposed that direct oral anticoagulants (DOACs) counteract Aβ-induced pathological alterations in cerebral blood vessels early in AD, a condition, known as cerebral amyloid angiopathy (CAA). By inhibiting thrombin for fibrin formation, anticoagulants can prevent accumulations of proinflammatory thrombin and fibrin, and deposition of degradation-resistant, Aβ-containing fibrin clots. These fibrin-Aβ clots are found in brain parenchyma between neuron cells, and in and around cerebral blood vessels in areas of CAA, leading to decreased cerebral blood flow. Consequently, anticoagulant treatment could reduce hypoperfusion and restricted supply of brain tissue with oxygen and nutrients. Concomitantly, hypoperfusion-enhanced neurodegenerative processes, such as progressive Aβ accumulation via synthesis and reduced perivascular clearance, neuroinflammation, and synapse and neuron cell loss, could be mitigated. Given full cerebral perfusion and reduced Aβ- and fibrin-accumulating and inflammatory milieu, anticoagulants could be able to decrease vascular-driven progression in neurodegenerative and cognitive changes, present in AD, when treated early, therapeutically, or prophylactically.
Collapse
Affiliation(s)
- Klaus Grossmann
- Center for Plant Molecular Biology (ZMBP), University of Tübingen, Tübingen, Germany
| |
Collapse
|
29
|
CZYH Alleviates β-Amyloid-Induced Cognitive Impairment and Inflammation Response via Modulation of JNK and NF- κB Pathway in Rats. Behav Neurol 2019; 2019:9546761. [PMID: 31781295 PMCID: PMC6875391 DOI: 10.1155/2019/9546761] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 08/17/2019] [Accepted: 09/09/2019] [Indexed: 11/17/2022] Open
Abstract
Cu-Zhi-Yi-Hao (CZYH), an empirical formula of traditional Chinese medicine (TCM), has been used for amnesia treatment in clinical practice. However, its underlying pharmacological mechanism has not been fully illuminated. The current study was designed to investigate the neuroprotective effect of CZYH on a β-amyloid 25-35- (Aβ25-35-) induced learning and memory deficit rat model. CZYH (200, 400, or 800 mg/kg), donepezil (1.0 mg/kg), or distilled water was given to Aβ25-35-stimulated animals for 17 days consecutively. The Morris water maze test revealed that CZYH (400 or 800 mg/kg) administration improved the Aβ25-35-induced cognitive impairments in rats, and Nissl staining demonstrated that CZYH mitigated the Aβ-caused neuron loss. In addition, CZYH treatment markedly inhibited the activation of microglia as evidenced by a decreased level of IBA-1 and increased YM-1/2 protein expression. The protein expression levels of TNF-α, IL-1β, and COX-2 were also repressed by CZYH. Besides, CZYH treatment alleviated Aβ-induced IκB-α degradation and NF-κB p65 phosphorylation, as well as reduced the JNK phosphorylation level. In conclusion, the present study suggests that CZYH could improve learning and memory abilities and relieve neuron loss in Aβ25-35-induced rats, at least partly through inhibition of the neuroinflammatory response via inhibiting the JNK-dependent NF-κB activation, indicating that CZYH might be a promising formula for the treatment of AD.
Collapse
|
30
|
Sun J, Ashley J, Kellawan JM. Can Acupuncture Treatment of Hypertension Improve Brain Health? A Mini Review. Front Aging Neurosci 2019; 11:240. [PMID: 31572163 PMCID: PMC6753179 DOI: 10.3389/fnagi.2019.00240] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 08/19/2019] [Indexed: 12/29/2022] Open
Abstract
With age, cerebrovascular and neurodegenerative diseases (e.g., dementia and Alzheimer’s) are some of the leading causes of death in the United States. Related to these outcomes is the increased prevalence of hypertension, which independently increases the development of cerebrovascular and neurodegenerative diseases. While a direct mechanistic link between hypertension and poor brain health is unknown, many hypothesize that the etiology stems from poor blood pressure (BP) and cerebrovascular regulation. This dysfunction fosters hypoperfusion of the brain, causing stress to the tissue through a nutrient mismatch, subtly damaging the brain over many years. Current Western medical treatment relies on pharmacological treatment (mainly beta-blockers, angiotensin-converting enzyme inhibitors, or a combination of the two). However, Western treatments have not been successful in mitigating brain health outcomes and are burdened with unwanted side effects and non-adherence issues. Alternatively, traditional East Asia medicine has used acupuncture as a treatment for hypertension and may offer a promising approach in response to the limitations of conventional therapy. While detailed clinical and mechanistic experimental evidence is lacking, acupuncture has been observed to reduce BP and improve endothelial function in hypertensive adults. Further, acupuncture has been shown to have specific cerebrovascular effects, increasing cerebrovascular reactivity in healthy adults, highlighting possible neuroprotective properties. Therefore, our review is aimed at evaluating acupuncture as a treatment for hypertension and the potential impact on brain health. We will interrogate the current literature as well as discuss the proposed neural and vascular mechanisms by which acupuncture acts.
Collapse
Affiliation(s)
- Jongjoo Sun
- Human Circulation Research Laboratory, Department of Health and Exercise Science, University of Oklahoma, Norman, OK, United States
| | - John Ashley
- Human Circulation Research Laboratory, Department of Health and Exercise Science, University of Oklahoma, Norman, OK, United States
| | - J Mikhail Kellawan
- Human Circulation Research Laboratory, Department of Health and Exercise Science, University of Oklahoma, Norman, OK, United States
| |
Collapse
|
31
|
Wang R, Wang C, Dai Z, Chen Y, Shen Z, Xiao G, Chen Y, Zhou JN, Zhuang Z, Wu R. An Amyloid-β Targeting Chemical Exchange Saturation Transfer Probe for In Vivo Detection of Alzheimer's Disease. ACS Chem Neurosci 2019; 10:3859-3867. [PMID: 31343167 DOI: 10.1021/acschemneuro.9b00334] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
A reliable and reproducible detection of Aβ deposits would be beneficial for the early diagnosis of Alzheimer's disease (AD). In the present study, the feasibility of applying chemical exchange saturation transfer (CEST) for Aβ deposit detection using angiopep-2 as a probe was evaluated, and it was demonstrated that CEST could detect angiopep-2 and Aβ-angiopep-2 aggregates in vitro. Furthermore, APP/PS1 mice injected with angiopep-2 exhibited a significantly higher in vivo CEST effect when compared with controls. The distribution of Aβ deposits detected by CEST imaging was consistent with the histological staining results. The present study is the first to report a reliable exogenous CEST probe to noninvasively evaluate Aβ deposits in APP/PS1 mice. Furthermore, these results demonstrate the potential for clinical AD diagnosis and Aβ-targeted drug therapy assessment using CEST imaging with the angiopep-2 probe.
Collapse
Affiliation(s)
- Runrun Wang
- Department of Medical Imaging, Second Affiliated Hospital , Shantou University Medical College , Shantou , Guangdong 515000 , P. R. China
| | - Chenwei Wang
- CAS Key Laboratory of Brain Function and Disease, School of Life Sciences , University of Science and Technology of China , Hefei , Anhui 230000 , P. R. China
| | - Zhuozhi Dai
- Department of Medical Imaging, Second Affiliated Hospital , Shantou University Medical College , Shantou , Guangdong 515000 , P. R. China
| | - Yanzi Chen
- Department of Medical Imaging, Second Affiliated Hospital , Shantou University Medical College , Shantou , Guangdong 515000 , P. R. China
| | - Zhiwei Shen
- Department of Medical Imaging, Second Affiliated Hospital , Shantou University Medical College , Shantou , Guangdong 515000 , P. R. China
| | - Gang Xiao
- Department of Mathematics and Statistics , Hanshan Normal University , Chaozhou 515000 , P. R. China
| | - Yuanfeng Chen
- Department of Medical Imaging, Second Affiliated Hospital , Shantou University Medical College , Shantou , Guangdong 515000 , P. R. China
| | - Jiang-Ning Zhou
- CAS Key Laboratory of Brain Function and Disease, School of Life Sciences , University of Science and Technology of China , Hefei , Anhui 230000 , P. R. China
- Center for Excellence in Brain Science and Intelligence Technology , Chinese Academy of Sciences , Shanghai 200031 , China
| | - Zerui Zhuang
- Department of Medical Imaging, Second Affiliated Hospital , Shantou University Medical College , Shantou , Guangdong 515000 , P. R. China
| | - Renhua Wu
- Department of Medical Imaging, Second Affiliated Hospital , Shantou University Medical College , Shantou , Guangdong 515000 , P. R. China
| |
Collapse
|
32
|
Neutrophil adhesion in brain capillaries reduces cortical blood flow and impairs memory function in Alzheimer's disease mouse models. Nat Neurosci 2019; 22:413-420. [PMID: 30742116 PMCID: PMC6508667 DOI: 10.1038/s41593-018-0329-4] [Citation(s) in RCA: 313] [Impact Index Per Article: 52.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 12/17/2018] [Indexed: 01/18/2023]
Abstract
Cerebral blood flow (CBF) reductions in Alzheimer’s disease (AD) patients and related mouse models have been recognized for decades, but the underlying mechanisms and resulting consequences on AD pathogenesis remain poorly understood. In APP/PS1 and 5xFAD mice we found that an increased number of cortical capillaries had stalled blood flow as compared to wildtype animals, largely due to neutrophils that adhered in capillary segments and blocked blood flow. Administration of antibodies against the neutrophil marker Ly6G reduced the number of stalled capillaries, leading to an immediate increase in CBF and to rapidly improved performance in spatial and working memory tasks. This study identified a novel cellular mechanism that explains the majority of the CBF reduction seen in two mouse models of AD and demonstrated that improving CBF rapidly improved short-term memory function. Restoring cerebral perfusion by preventing neutrophil adhesion may provide a novel strategy for improving cognition in AD patients.
Collapse
|
33
|
Alegría-Herrera E, Herrera-Ruiz M, Román-Ramos R, Zamilpa A, Santillán-Urquiza MA, Aguilar MI, Avilés-Flores M, Fuentes-Mata M, Jiménez-Ferrer E. Effect of Ocimum basilicum, Ocimum selloi, and Rosmarinic Acid on Cerebral Vascular Damage in a Chronic Hypertension Model. Biol Pharm Bull 2019; 42:201-211. [PMID: 30713252 DOI: 10.1248/bpb.b18-00574] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The main objective of treatment against hypertension is not only to reduce blood pressure levels, but also to reduce vascular risk in general. In the present work, administering angiotensin II (AGII; 0.2 µg/kg intraperitoneally (i.p.) for 12 weeks) activates the hypothalamic-pituitary-adrenal (HPA) axis, which caused an increase in corticosterone levels, as well as in proinflammatory cytokines (interleukin 1β (IL-1β), interleukin 6 (IL-6), and tumor necrosis factor alpha (TNF-α)) and macrophage chemotactic protein 1 (MCP-1), and decreased anti-inflammatory cytokines (interleukin 10 (IL-10) and interleukin 4 (IL-4)). On observing the behavior in the different models, an anxiogenic effect (elevated plus maze (EPM)) and cognitive impairment (water Morris maze (WMM)) was observed in animals with AGII. By administering organic extracts from Ocimum basilicum (Oba-EtOAc) and Ocimum selloi (Ose-EtOAc), and some doses of rosmarinic acid (RA) (6 weeks per os (p.o.)), the damage caused by AGII was stopped by re-establishing corticosterone serum levels and by decreasing the proinflammatory cytokines and MCP-1.
Collapse
Affiliation(s)
- Elian Alegría-Herrera
- Centro de Investigación Biomédica del Sur, Instituto Mexicano del Seguro Social (IMSS)
- Doctorado en Ciencias Biológicas y de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Iztapalapa
- Departamento Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Iztapalapa
| | - Maribel Herrera-Ruiz
- Centro de Investigación Biomédica del Sur, Instituto Mexicano del Seguro Social (IMSS)
| | - Rubén Román-Ramos
- Departamento Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Iztapalapa
| | - Alejandro Zamilpa
- Centro de Investigación Biomédica del Sur, Instituto Mexicano del Seguro Social (IMSS)
| | | | - María Isabel Aguilar
- Laboratorio 111, Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México
| | | | | | | |
Collapse
|
34
|
Abstract
PURPOSE OF REVIEW The extrinsic risk factors for postoperative cognitive disturbance have been a source of concern during the perioperative period, and these risk factors remain the subject of controversy. This review of recent studies focuses on the effect of these factors on postoperative cognitive disturbance during the perioperative period. RECENT FINDINGS Impairment of cerebral autoregulation may predispose patients to intraoperative cerebral malperfusion, which may subsequently induce postoperative cognitive disturbance. The neurotoxicity of several volatile anesthetics may contribute to cognitive functional decline, and the impact of intravenous anesthesia on cognitive function requires further exploration. Multimodal analgesia may not outperform traditional postoperative analgesia in preventing postoperative delirium. Furthermore, acute pain and chronic pain may exacerbate the cognitive functional decline of patients with preexisting cognitive impairment. The nuclear factor-kappa beta pathway is an important node in the neuroinflammatory network. SUMMARY Several intraoperative factors are associated with postoperative cognitive disturbance. However, if these factors are optimized in perioperative management, postoperative cognitive disturbance will improve.
Collapse
Affiliation(s)
- Huiqun Fu
- Department of Anesthesiology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | | | | |
Collapse
|
35
|
Tolomeo D, Micotti E, Serra SC, Chappell M, Snellman A, Forloni G. Chemical exchange saturation transfer MRI shows low cerebral 2-deoxy-D-glucose uptake in a model of Alzheimer's Disease. Sci Rep 2018; 8:9576. [PMID: 29934551 PMCID: PMC6015016 DOI: 10.1038/s41598-018-27839-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 06/11/2018] [Indexed: 12/17/2022] Open
Abstract
Glucose is the central nervous system's only energy source. Imaging techniques capable to detect pathological alterations of the brain metabolism are useful in different diagnostic processes. Such techniques are also beneficial for assessing the evaluation efficacy of therapies in pre-clinical and clinical stages of diseases. Chemical exchange saturation transfer (CEST) magnetic resonance imaging (MRI) is a possible alternative to positron emission tomography (PET) imaging that has been widely explored in cancer research in humans and animal models. We propose that pathological alterations in brain 2-deoxy-D-glucose (2DG) uptake, typical of neurodegenerative diseases, can be detected with CEST MRI. Transgenic mice overexpressing a mutated form of amyloid precusrsor protein (APP23), a model of Alzheimer's disease, analyzed with CEST MRI showed a clear reduction of 2DG uptake in different brain regions. This was reminiscent of the cerebral condition observed in Alzheimer's patients. The results indicate the feasibility of CEST for analyzing the brain metabolic state, with better image resolution than PET in experimental models.
Collapse
Affiliation(s)
- Daniele Tolomeo
- Laboratory of Biology of Neurodegenerative Disorders, Department of Neuroscience, IRCCS, Mario Negri Institute for Pharmacological Research, Milan, (MI), Italy
| | - Edoardo Micotti
- Laboratory of Biology of Neurodegenerative Disorders, Department of Neuroscience, IRCCS, Mario Negri Institute for Pharmacological Research, Milan, (MI), Italy
| | | | - Michael Chappell
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, 6396, Oxford, UK
| | - Anniina Snellman
- Medicity Research Laboratory, University of Turku, (Tykistökatu 6, FI-20510), Turku, Finland.,Turku PET Centre, University of Turku, (Kiinamyllynkatu 4-8, FI-20520,), Turku, Finland
| | - Gianluigi Forloni
- Laboratory of Biology of Neurodegenerative Disorders, Department of Neuroscience, IRCCS, Mario Negri Institute for Pharmacological Research, Milan, (MI), Italy.
| |
Collapse
|
36
|
Eguchi K, Shindo T, Ito K, Ogata T, Kurosawa R, Kagaya Y, Monma Y, Ichijo S, Kasukabe S, Miyata S, Yoshikawa T, Yanai K, Taki H, Kanai H, Osumi N, Shimokawa H. Whole-brain low-intensity pulsed ultrasound therapy markedly improves cognitive dysfunctions in mouse models of dementia - Crucial roles of endothelial nitric oxide synthase. Brain Stimul 2018; 11:959-973. [PMID: 29857968 DOI: 10.1016/j.brs.2018.05.012] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 05/01/2018] [Accepted: 05/20/2018] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Therapeutic focused-ultrasound to the hippocampus has been reported to exert neuroprotective effects on dementia. In the present study, we examined whether the whole-brain LIPUS (low-intensity pulsed ultrasound) therapy is effective and safe in 2 mouse models of dementia (vascular dementia, VaD and Alzheimer's disease, AD), and if so, to elucidate the common underlying mechanism(s) involved. METHODS We used bilateral carotid artery stenosis (BCAS) model with micro-coils in male C57BL/6 mice as a VaD model and 5XFAD transgenic mice as an AD model. We applied the LIPUS therapy (1.875 MHz, 6.0 kHz, 32cycles) to the whole brain. RESULTS In both models, the LIPUS therapy markedly ameliorated cognitive impairments (Y-maze test and/or passive avoidance test) associated with improved cerebral blood flow (CBF). Mechanistically, the LIPUS therapy significantly increased CD31-positive endothelial cells and Olig2-positive oligodendrocyte precursor cells (OPCs) in the VaD model, while it reduced Iba-1-positive microglias and amyloid-β (Aβ) plaque in the AD model. In both models, endothelium-related genes were significantly upregulated in RNA-sequencing, and expressions of endothelial nitric oxide synthase (eNOS) and neurotrophins were upregulated in Western blotting. Interestingly, the increases in glia cells and neurotrophin expressions showed significant correlations with eNOS expression. Importantly, these beneficial effects of LIPUS were absent in eNOS-knockout mice. CONCLUSIONS These results indicate that the whole-brain LIPUS is an effective and non-invasive therapy for dementia by activating specific cells corresponding to each pathology, for which eNOS activation plays an important role as a common mechanism.
Collapse
Affiliation(s)
- Kumiko Eguchi
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tomohiko Shindo
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kenta Ito
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tsuyoshi Ogata
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Ryo Kurosawa
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yuta Kagaya
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yuto Monma
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Sadamitsu Ichijo
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Sachie Kasukabe
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Satoshi Miyata
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takeo Yoshikawa
- Department of Pharmacology, Tohoku University School of Medicine Sendai, Japan
| | - Kazuhiko Yanai
- Department of Pharmacology, Tohoku University School of Medicine Sendai, Japan
| | - Hirofumi Taki
- Biomedical Engineering for Health and Welfare, Graduate School of Biomedical Engineering, Tohoku University, Sendai, Japan
| | - Hiroshi Kanai
- Department of Electronic Engineering, Tohoku University Graduate School of Engineering, Sendai, Japan
| | - Noriko Osumi
- Department of Developmental Neuroscience, Tohoku University, Sendai, Japan
| | - Hiroaki Shimokawa
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan.
| |
Collapse
|
37
|
Asaad M, Lee JH. A guide to using functional magnetic resonance imaging to study Alzheimer's disease in animal models. Dis Model Mech 2018; 11:dmm031724. [PMID: 29784664 PMCID: PMC5992611 DOI: 10.1242/dmm.031724] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Alzheimer's disease is a leading healthcare challenge facing our society today. Functional magnetic resonance imaging (fMRI) of the brain has played an important role in our efforts to understand how Alzheimer's disease alters brain function. Using fMRI in animal models of Alzheimer's disease has the potential to provide us with a more comprehensive understanding of the observations made in human clinical fMRI studies. However, using fMRI in animal models of Alzheimer's disease presents some unique challenges. Here, we highlight some of these challenges and discuss potential solutions for researchers interested in performing fMRI in animal models. First, we briefly summarize our current understanding of Alzheimer's disease from a mechanistic standpoint. We then overview the wide array of animal models available for studying this disease and how to choose the most appropriate model to study, depending on which aspects of the condition researchers seek to investigate. Finally, we discuss the contributions of fMRI to our understanding of Alzheimer's disease and the issues to consider when designing fMRI studies for animal models, such as differences in brain activity based on anesthetic choice and ways to interrogate more specific questions in rodents beyond those that can be addressed in humans. The goal of this article is to provide information on the utility of fMRI, and approaches to consider when using fMRI, for studies of Alzheimer's disease in animal models.
Collapse
Affiliation(s)
- Mazen Asaad
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA 94305, USA
| | - Jin Hyung Lee
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
- Department of Electrical Engineering, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
38
|
Zhang C, Feng W, Zhao Y, Yu T, Li P, Xu T, Luo Q, Zhu D. A large, switchable optical clearing skull window for cerebrovascular imaging. Theranostics 2018; 8:2696-2708. [PMID: 29774069 PMCID: PMC5957003 DOI: 10.7150/thno.23686] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 03/01/2018] [Indexed: 12/19/2022] Open
Abstract
Rationale: Intravital optical imaging is a significant method for investigating cerebrovascular structure and function. However, its imaging contrast and depth are limited by the turbid skull. Tissue optical clearing has a great potential for solving this problem. Our goal was to develop a transparent skull window, without performing a craniotomy, for use in assessing cerebrovascular structure and function. Methods: Skull optical clearing agents were topically applied to the skulls of mice to create a transparent window within 15 min. The clearing efficacy, repeatability, and safety of the skull window were then investigated. Results: Imaging through the optical clearing skull window enhanced both the contrast and the depth of intravital imaging. The skull window could be used on 2-8-month-old mice and could be expanded from regional to bi-hemispheric. In addition, the window could be repeatedly established without inducing observable inflammation and metabolic toxicity. Conclusion: We successfully developed an easy-to-handle, large, switchable, and safe optical clearing skull window. Combined with various optical imaging techniques, cerebrovascular structure and function can be observed through this optical clearing skull window. Thus, it has the potential for use in basic research on the physiopathologic processes of cortical vessels.
Collapse
Affiliation(s)
- Chao Zhang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
- MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Wei Feng
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
- MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Yanjie Zhao
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
- MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Tingting Yu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
- MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Pengcheng Li
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
- MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Tonghui Xu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
- MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Qingming Luo
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
- MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Dan Zhu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
- MoE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| |
Collapse
|