1
|
Thangwong P, Tocharus C, Tocharus J. The Bidirectional Role of Hypoxia-Inducible Factor 1 Alpha in Vascular Dementia Caused by Chronic Cerebral Hypoperfusion. Mol Neurobiol 2025:10.1007/s12035-025-04914-5. [PMID: 40205304 DOI: 10.1007/s12035-025-04914-5] [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: 10/25/2024] [Accepted: 04/03/2025] [Indexed: 04/11/2025]
Abstract
Chronic cerebral hypoperfusion (CCH) is a critical indicator of cognitive impairment and dementia, especially vascular dementia. Cerebral blood flow disturbance alters the properties of neurons and glial cells as a result of a deficit in energy sources. Hypoxia-inducible factor 1 alpha (HIF- 1α) is a transcription factor that controls gene activity in response to low oxygen levels. It regulates a complex network of cellular adaptations to improve oxygenation, metabolic reprogramming, and cell survival in hypoxic situations. However, recent research suggests that HIF- 1α plays a role not only in neuroprotection but also in brain injury. It is therefore critical to fully comprehend the mechanisms behind these disorders. This review highlights the dual role of HIF- 1α in CCH-induced VaD. Initially, HIF- 1α provides a neuroprotection by promoting angiogenesis through vascular endothelial growth factor (VEGF) signaling. However, prolonged activation can detrimentally effects, including oxidative stress, neuroinflammation, blood-brain barrier dysfunction, and cognitive impairment. Evidence suggests that HIF- 1α exerts its protective effects in acute ischemic/hypoxic-induced VaD through pathways such as PI3 K/AKT/mTOR and MAPK/p-c-Jun signaling. However, its dysregulation in chronic stages of CCH contributes to cognitive decline and disease progression. Understanding the complex role of HIF- 1α and its interactions with other molecular pathways is crucial for developing effective therapeutic strategies. Therefore, an informed, in-depth discussion of its involvement in these pathologic processes is necessary, as a precise contribution of HIF- 1α to CCH-induced VaD remains to be established and requires further investigation.
Collapse
Affiliation(s)
- Phakkawat Thangwong
- Department of Medical Science, School of Medicine, Walailak University, Nakhon Si Thammarat, 80160, Thailand
- Research Center in Tropical Pathobiology, Walailak University, Nakhon Si Thammarat, 80160, Thailand
| | - Chainarong Tocharus
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Functional Food Research Center for Well-Being, Multidisciplinary Research Institute, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Jiraporn Tocharus
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.
- Functional Food Research Center for Well-Being, Multidisciplinary Research Institute, Chiang Mai University, Chiang Mai, 50200, Thailand.
| |
Collapse
|
2
|
Zahra S, Rubab Khakwani KZ, Acosta D, Arias JC, French S, Bedrick EJ, Vitali F, Beach TG, Serrano G, Weinkauf CC. Neurofibrillary tangles predict dementia in patients with carotid stenosis. J Vasc Surg 2025:S0741-5214(25)00330-1. [PMID: 40010674 DOI: 10.1016/j.jvs.2025.02.007] [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: 11/27/2024] [Revised: 01/31/2025] [Accepted: 02/05/2025] [Indexed: 02/28/2025]
Abstract
OBJECTIVE There is growing appreciation that extracranial carotid atherosclerotic disease (ECAD) is associated with increased dementia risk. Despite this, clinical management of ECAD does not involve evaluation for cognitive outcomes or risk stratification for dementia. One impediment to studying and improving clinical care for this cohort (roughly 10% of adults aged >60 years) is that factors to identify patients with ECAD at risk for dementia are not known. METHODS Our prospective clinicopathologic study, the Arizona Study of Aging and Neurodegenerative Disorders study, evaluated clinical and histopathologic factors for dementia in subjects with ECAD. The primary outcome (dementia) was defined as a composite of Alzheimer's disease and/or vascular dementia based on a clinical/neuropathologic diagnosis. Of 1234 subjects, those with dementia other than Alzheimer's disease and/or vascular dementia were excluded; there remained 111 subjects with ECAD to be evaluated. Logistic regression analysis was performed to examine the association of key risk factors for dementia including age, sex, cardiovascular risk factors, apolipoprotein E4 (APOE4) genetic status, and dementia biomarkers. A precision recall curve was also generated to evaluate the diagnostic accuracy of dementia prediction models. RESULTS Individuals with dementia compared with those without had significantly increased levels of stroke, APOE4 genotype, and dementia biomarkers, neurofibrillary tangles (NFTs), and amyloid plaques. Models of multiple combined risk factors were little or no better than NFTs alone, which showed a 96.9% positive predictive value at an NFT threshold of 10. CONCLUSIONS Although we hypothesized that a combination of clinical and histopathologic biomarkers would result in the strongest predictive model for dementia, we found that NFTs alone had the highest association and positive predictive value for dementia risk in patients with ECAD. As blood-based assays for NFT quantification become more clinically reliable and available, these data support the possibility that NFT quantification may help identify patients with ECAD at increased risk for dementia.
Collapse
Affiliation(s)
- Summan Zahra
- Department of Surgery, University of Arizona College of Medicine, Tucson, AZ
| | | | - Daniela Acosta
- Department of Surgery, University of Arizona College of Medicine, Tucson, AZ
| | - Juan C Arias
- Department of Surgery, University of Arizona College of Medicine, Tucson, AZ
| | - Scott French
- Department of Surgery, University of Arizona College of Medicine, Tucson, AZ
| | - Edward J Bedrick
- Department of Epidemiology and Biostatistics, University of Arizona College of Public Health, Tucson, AZ
| | - Francesca Vitali
- Center for Innovation in Brain Science, Department of Neurology, University of Arizona College of Medicine, Tucson, AZ
| | - Thomas G Beach
- Brain and Body Donation Program, Banner Sun Health Research Institute, Sun City, AZ
| | - Geidy Serrano
- Brain and Body Donation Program, Banner Sun Health Research Institute, Sun City, AZ
| | - Craig C Weinkauf
- Department of Surgery, University of Arizona College of Medicine, Tucson, AZ.
| |
Collapse
|
3
|
Lee NK, Na DL, Kim HJ, Jang H, Sa JK, Ko BS, Chang JW. Prolonged Chronic Cerebral Hypoperfusion Does not Exacerbate Tau Pathology in a Tauopathy Mouse Model. J Integr Neurosci 2025; 24:26108. [PMID: 40018776 DOI: 10.31083/jin26108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 10/20/2024] [Accepted: 10/25/2024] [Indexed: 03/01/2025] Open
Abstract
BACKGROUND Several preclinical studies have reported elevated levels of tau following the induction of chronic cerebral hypoperfusion (CCH) in Alzheimer's disease mouse models. The objective of this study was to first induce CCH in a mouse model of tauopathy over an extended period of up to 6 months and to subsequently investigate the effects of CCH on tau accumulation and alterations in the transcriptome. METHODS Three-month-old P301S tauopathy mice were randomly allocated to either a Sham or CCH group. The common carotid arteries (CCAs) of the CCH group were bilaterally implanted using 0.75-mm inner diameter ameroid constrictors. Prior to surgery, Doppler ultrasound imaging was acquired, with follow-up imaging at 1, 3, and 6 months postoperatively. Brain tissue samples were obtained, and hemispheres were dissected and divided for separate analysis. RESULT No significant differences in phosphorylated and total tau protein levels were found in either Sham or CCH left cortical hemispheres or hippocampal lysates. Immunohistochemical staining of phosphorylated tau in the right hemisphere revealed similar findings. Compared with the Sham group, transcriptomic deconvolution revealed a significant reduction of memory B cells in the CCH group (p = 0.029). CONCLUSION To clarify the effects of chronic hypoperfusion on tau pathology, more than one surgical method of hypoperfusion should be used in future studies.
Collapse
Affiliation(s)
- Na Kyung Lee
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, 06355 Seoul, Republic of Korea
- Cell and Gene Therapy Institute (CGTI), Research Institute for Future Medicine, Samsung Medical Center, 06351 Seoul, Republic of Korea
| | - Duk L Na
- Cell and Gene Therapy Institute (CGTI), Research Institute for Future Medicine, Samsung Medical Center, 06351 Seoul, Republic of Korea
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, 06351 Seoul, Republic of Korea
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 06351 Seoul, Republic of Korea
- Happymind Clinic, 06061 Seoul, Republic of Korea
| | - Hee Jin Kim
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, 06355 Seoul, Republic of Korea
- Cell and Gene Therapy Institute (CGTI), Research Institute for Future Medicine, Samsung Medical Center, 06351 Seoul, Republic of Korea
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, 06351 Seoul, Republic of Korea
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 06351 Seoul, Republic of Korea
- Department of Digital Health, SAIHST, Sungkyunkwan University, 06355 Seoul, Republic of Korea
| | - Hyemin Jang
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, 03080 Seoul, Republic of Korea
| | - Jason K Sa
- Department of Biomedical Informatics, Korea University College of Medicine, 02841 Seoul, Republic of Korea
| | - Bae Sung Ko
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, 06355 Seoul, Republic of Korea
- Cell and Gene Therapy Institute (CGTI), Research Institute for Future Medicine, Samsung Medical Center, 06351 Seoul, Republic of Korea
| | - Jong Wook Chang
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, 06355 Seoul, Republic of Korea
- Cell and Gene Therapy Institute (CGTI), Research Institute for Future Medicine, Samsung Medical Center, 06351 Seoul, Republic of Korea
- Cell & Gene Therapy Research Institute, ENCell Co., Ltd., 06072 Seoul, Republic of Korea
| |
Collapse
|
4
|
Gheni G, Shinohara M, Masuda‐Suzukake M, Shindo A, Watanabe A, Kawai K, Bu G, Tomimoto H, Hasegawa M, Sato N. Cerebral hypoperfusion reduces tau accumulation. Ann Clin Transl Neurol 2025; 12:69-85. [PMID: 39621511 PMCID: PMC11752094 DOI: 10.1002/acn3.52247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 10/03/2024] [Accepted: 10/24/2024] [Indexed: 01/22/2025] Open
Abstract
OBJECTIVE Alzheimer's disease (AD) often coexists with cerebrovascular diseases. However, the impact of cerebrovascular diseases such as stroke on AD pathology remains poorly understood. METHODS This study examines the correlation between cerebrovascular diseases and AD pathology. The research was carried out using clinical and neuropathological data collected from the National Alzheimer's Coordinating Center (NACC) database and an animal model in which bilateral common carotid artery stenosis surgery was performed, following the injection of tau seeds into the brains of wild-type mice. RESULTS Analysis of the NACC database suggests that clinical stroke history and lacunar infarcts are associated with lower neurofibrillary tangle pathology. An animal model demonstrates that chronic cerebral hypoperfusion reduces tau pathology, which was observed in not only neurons but also astrocytes, microglia, and oligodendrocytes. Furthermore, we found that astrocytes and microglia were activated in response to tau pathology and chronic cerebral hypoperfusion. Additionally, cerebral hypoperfusion increased a lysosomal enzyme, cathepsin D. INTERPRETATION These data together indicate that cerebral hypoperfusion reduces tau accumulation likely through an increase in microglial phagocytic activity towards tau and an elevation in degradation through cathepsin D. This study contributes to understanding the relationship between tau pathology and cerebrovascular diseases in older people with multimorbidity.
Collapse
Grants
- MEXT15K15272 Grants-in-Aid from Japan Promotion of Science; the Japanese Ministry of Education, Culture, Sports, Science, and Technology
- 19-9 Funding for Longevity Sciences from the National Center for Geriatrics and Gerontology
- P20 AG068053 NIA NIH HHS
- P30 AG062421 NIA NIH HHS
- Takeda Medical Research Foundation Research
- P30 AG066508 NIA NIH HHS
- P30 AG072973 NIA NIH HHS
- P30 AG066530 NIA NIH HHS
- Novartis Foundation for Gerontological Research Award
- P30 AG066509 NIA NIH HHS
- P30 AG066546 NIA NIH HHS
- Mitsui Sumitomo Insurance Welfare Foundation
- P30 AG072979 NIA NIH HHS
- P20 AG068082 NIA NIH HHS
- P30 AG072975 NIA NIH HHS
- P30 AG066444 NIA NIH HHS
- P30 AG066507 NIA NIH HHS
- P30 AG072946 NIA NIH HHS
- P30 AG066518 NIA NIH HHS
- P30 AG066511 NIA NIH HHS
- U24 AG072122 NIA NIH HHS
- MEXT21H02844 Grants-in-Aid from Japan Promotion of Science; the Japanese Ministry of Education, Culture, Sports, Science, and Technology
- P30 AG066512 NIA NIH HHS
- MEXT26293167 Grants-in-Aid from Japan Promotion of Science; the Japanese Ministry of Education, Culture, Sports, Science, and Technology
- P30 AG066515 NIA NIH HHS
- P30 AG072978 NIA NIH HHS
- P30 AG062429 NIA NIH HHS
- P30 AG066519 NIA NIH HHS
- Takeda Science Foundation Research Encouragement Grant
- 28-45 Funding for Longevity Sciences from the National Center for Geriatrics and Gerontology
- P30 AG062422 NIA NIH HHS
- R01 AG079280 NIA NIH HHS
- P30 AG066462 NIA NIH HHS
- 19-3 Funding for Longevity Sciences from the National Center for Geriatrics and Gerontology
- 21-12 Funding for Longevity Sciences from the National Center for Geriatrics and Gerontology
- MEXT24K02361 Grants-in-Aid from Japan Promotion of Science; the Japanese Ministry of Education, Culture, Sports, Science, and Technology
- P20 AG068077 NIA NIH HHS
- P30 AG072977 NIA NIH HHS
- P30 AG062677 NIA NIH HHS
- P20 AG068024 NIA NIH HHS
- P30 AG072958 NIA NIH HHS
- P30 AG062715 NIA NIH HHS
- P30 AG066506 NIA NIH HHS
- P30 AG066468 NIA NIH HHS
- Annual Research Award Grant from the Japanese Society of Anti-aging Medicine
- P30 AG072976 NIA NIH HHS
- P30 AG072947 NIA NIH HHS
- P30 AG072931 NIA NIH HHS
- MEXT17H04154 Grants-in-Aid from Japan Promotion of Science; the Japanese Ministry of Education, Culture, Sports, Science, and Technology
- SENSHIN Medical Research Foundation Research Grant
- P30 AG072972 NIA NIH HHS
- P30 AG066514 NIA NIH HHS
- P30 AG072959 NIA NIH HHS
- 24-16 Funding for Longevity Sciences from the National Center for Geriatrics and Gerontology
- Mitsui Sumitomo Insurance Welfare Foundation
Collapse
Affiliation(s)
- Ghupurjan Gheni
- Department of Aging Neurobiology, Center for Development of Advanced Medicine for DementiaNational Center for Geriatrics and Gerontology7‐430 MoriokaObuAichi474‐8511Japan
| | - Mitsuru Shinohara
- Department of Aging Neurobiology, Center for Development of Advanced Medicine for DementiaNational Center for Geriatrics and Gerontology7‐430 MoriokaObuAichi474‐8511Japan
- Department of NeuroscienceMayo ClinicJacksonvilleFlorida32224USA
| | - Masami Masuda‐Suzukake
- Dementia Research ProjectTokyo Metropolitan Institute of Medical Science2‐1‐6 Kamikitazawa, Setagaya‐kuTokyo156‐8506Japan
| | - Akihiko Shindo
- Department of Neurology, Graduate School of MedicineMie University1577 Kurima MachiyachoTsu CityMie514‐8507Japan
| | - Atsushi Watanabe
- Equipment Management Division, Center for Core Facility Administration, National Center for Geriatrics and Gerontology7‐430 MoriokaObuAichi474‐8511Japan
| | - Kaori Kawai
- Department of Aging Neurobiology, Center for Development of Advanced Medicine for DementiaNational Center for Geriatrics and Gerontology7‐430 MoriokaObuAichi474‐8511Japan
| | - Guojun Bu
- Department of NeuroscienceMayo ClinicJacksonvilleFlorida32224USA
- Division of Life ScienceThe Hong Kong University of Science and TechnologyHong KongChina
| | - Hidekazu Tomimoto
- Department of Neurology, Graduate School of MedicineMie University1577 Kurima MachiyachoTsu CityMie514‐8507Japan
| | - Masato Hasegawa
- Dementia Research ProjectTokyo Metropolitan Institute of Medical Science2‐1‐6 Kamikitazawa, Setagaya‐kuTokyo156‐8506Japan
| | - Naoyuki Sato
- Department of Aging Neurobiology, Center for Development of Advanced Medicine for DementiaNational Center for Geriatrics and Gerontology7‐430 MoriokaObuAichi474‐8511Japan
| |
Collapse
|
5
|
Lee H, Fu JF, Gaudet K, Bryant AG, Price JC, Bennett RE, Johnson KA, Hyman BT, Hedden T, Salat DH, Yen YF, Huang SY. Aberrant vascular architecture in the hippocampus correlates with tau burden in mild cognitive impairment and Alzheimer's disease. J Cereb Blood Flow Metab 2024; 44:787-800. [PMID: 38000018 PMCID: PMC11197134 DOI: 10.1177/0271678x231216144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 09/04/2023] [Accepted: 10/21/2023] [Indexed: 11/26/2023]
Abstract
Cerebrovascular dysfunction is a significant contributor to Alzheimer's disease (AD) progression. AD mouse models show altered capillary morphology, density, and diminished blood flow in areas of tau and beta-amyloid accumulation. The purpose of this study was to examine alterations in vascular structure and their contributions to perfusion deficits in the hippocampus in AD and mild cognitive impairment (MCI). Seven individuals with AD and MCI (1 AD/6 MCI), nine cognitively intact older healthy adults, and seven younger healthy adults underwent pseudo-continuous arterial spin labeling (PCASL) and gradient-echo/spin-echo (GESE) dynamic susceptibility contrast (DSC) MRI. Cerebral blood flow (CBF), cerebral blood volume, relative vessel size index (rVSI), and mean vessel density were calculated from model fitting. Lower CBF from PCASL and SE DSC MRI was observed in the hippocampus of AD/MCI group. rVSI in the hippocampus of the AD/MCI group was larger than that of the two healthy groups (FDR-P = 0.02). No difference in vessel density was detected between the groups. We also explored relationship of tau burden from 18F-flortaucipir positron emission tomography and vascular measures from MRI. Tau burden was associated with larger vessel size and lower CBF in the hippocampus. We postulate that larger vessel size may be associated with vascular alterations in AD/MCI.
Collapse
Affiliation(s)
- Hansol Lee
- Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology, Ulsan, South Korea
| | - Jessie Fanglu Fu
- Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Kyla Gaudet
- Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Annie G Bryant
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Julie C Price
- Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Rachel E Bennett
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Keith A Johnson
- Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital, Boston, MA, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | - Bradley T Hyman
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Trey Hedden
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - David H Salat
- Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Yi-Fen Yen
- Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Susie Y Huang
- Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
| |
Collapse
|
6
|
Fu P, Chen Y, Wu M, Bao B, Yin X, Chen Z, Zhang M. Effect of ferroptosis on chronic cerebral hypoperfusion in vascular dementia. Exp Neurol 2023; 370:114538. [PMID: 37709116 DOI: 10.1016/j.expneurol.2023.114538] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 08/22/2023] [Accepted: 09/10/2023] [Indexed: 09/16/2023]
Abstract
Vascular dementia (VaD) is the second most prevalent type of dementia after Alzheimer's disease and is caused by impaired nerve cell function resulting from cerebrovascular disease and vascular risk factors. Chronic cerebral hypoperfusion (CCH) is a common pathological and physiological state that may result from cerebral ischemia and hypoxia, causing widespread diffuse lesions in the brain parenchyma which leads to progressive nerve damage. Transferrin (TF) and transferrin receptor 1 (TfR1), two proteins involved in iron uptake, were upregulated by CCH, whereas ferroprotein (FPN), a protein involved in iron efflux, was downregulated. This process may involve various mechanisms including tau and iron regulatory proteins (IRP). CCH can also exacerbate lipid peroxidation caused by an iron imbalance by inhibiting glutathione peroxidase 4 (Gpx4) synthesis and some Gpx4 independent pathways through cystine/glutamate transporters (system Xc-), ultimately leading to ferroptosis in nerve cells and accelerating the progression of VaD.
Collapse
Affiliation(s)
- Peijie Fu
- Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi 332000, China; Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China
| | - Yanghang Chen
- Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi 332000, China; Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China; Department of Neurology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, China
| | - Moxin Wu
- Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi 332000, China; Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China
| | - Bing Bao
- Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi 332000, China; Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China
| | - Xiaoping Yin
- Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi 332000, China; Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China
| | - Zhiying Chen
- Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi 332000, China; Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China.
| | - Manqing Zhang
- School of Basic Medicine, Jiujiang University, Jiujiang, Jiangxi 332000, China.
| |
Collapse
|
7
|
Ishikawa H, Shindo A, Mizutani A, Tomimoto H, Lo EH, Arai K. A brief overview of a mouse model of cerebral hypoperfusion by bilateral carotid artery stenosis. J Cereb Blood Flow Metab 2023; 43:18-36. [PMID: 36883344 PMCID: PMC10638994 DOI: 10.1177/0271678x231154597] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 12/23/2022] [Accepted: 01/04/2023] [Indexed: 03/09/2023]
Abstract
Vascular cognitive impairment (VCI) refers to all forms of cognitive disorder related to cerebrovascular diseases, including vascular mild cognitive impairment, post-stroke dementia, multi-infarct dementia, subcortical ischemic vascular dementia (SIVD), and mixed dementia. Among the causes of VCI, more attention has been paid to SIVD because the causative cerebral small vessel pathologies are frequently observed in elderly people and because the gradual progression of cognitive decline often mimics Alzheimer's disease. In most cases, small vessel diseases are accompanied by cerebral hypoperfusion. In mice, prolonged cerebral hypoperfusion is induced by bilateral carotid artery stenosis (BCAS) with surgically implanted metal micro-coils. This cerebral hypoperfusion BCAS model was proposed as a SIVD mouse model in 2004, and the spreading use of this mouse SIVD model has provided novel data regarding cognitive dysfunction and histological/genetic changes by cerebral hypoperfusion. Oxidative stress, microvascular injury, excitotoxicity, blood-brain barrier dysfunction, and secondary inflammation may be the main mechanisms of brain damage due to prolonged cerebral hypoperfusion, and some potential therapeutic targets for SIVD have been proposed by using transgenic mice or clinically used drugs in BCAS studies. This review article overviews findings from the studies that used this hypoperfused-SIVD mouse model, which were published between 2004 and 2021.
Collapse
Affiliation(s)
- Hidehiro Ishikawa
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Akihiro Shindo
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Akane Mizutani
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Hidekazu Tomimoto
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Eng H Lo
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Ken Arai
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| |
Collapse
|
8
|
Ahmadi K, Pereira JB, Berron D, Vogel J, Ingala S, Strandberg OT, Janelidze S, Barkhof F, Pfeuffer J, Knutsson L, van Westen D, Palmqvist S, Mutsaerts HJ, Hansson O. Gray matter hypoperfusion is a late pathological event in the course of Alzheimer's disease. J Cereb Blood Flow Metab 2023; 43:565-580. [PMID: 36412244 PMCID: PMC10063832 DOI: 10.1177/0271678x221141139] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Several studies have shown decreased cerebral blood flow (CBF) in Alzheimer's disease (AD). However, the role of hypoperfusion in the disease pathogenesis remains unclear. Combining arterial spin labeling MRI, PET, and CSF biomarkers, we investigated the associations between gray matter (GM)-CBF and the key mechanisms in AD including amyloid-β (Aβ) and tau pathology, synaptic and axonal degeneration. Further, we applied a disease progression modeling to characterize the temporal sequence of different AD biomarkers. Lower perfusion was observed in temporo-occipito-parietal cortex in the Aβ-positive cognitively impaired compared to both Aβ-negative and Aβ-positive cognitively unimpaired individuals. In participants along the AD spectrum, GM-CBF was associated with tau, synaptic and axonal dysfunction, but not Aβ in similar cortical regions. Axonal degeneration was further associated with hypoperfusion in cognitively unimpaired individuals. Disease progression modeling revealed that GM-CBF disruption Followed the abnormality of biomarkers of Aβ, tau and brain atrophy. These findings indicate that tau tangles and neurodegeneration are more closely connected with GM-CBF changes than Aβ pathology. Although subjected to the sensitivity of the employed neuroimaging techniques and the modeling approach, these findings suggest that hypoperfusion might not be an early event associated with the build-up of Aβ in preclinical phase of AD.
Collapse
Affiliation(s)
- Khazar Ahmadi
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden.,Institute of Cognitive Neuroscience, Faculty of Psychology, Ruhr University Bochum, Bochum, Germany
| | - Joana B Pereira
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden.,Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm, Sweden
| | - David Berron
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Jacob Vogel
- Department of Psychiatry, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Silvia Ingala
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Location VUmc, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Olof T Strandberg
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Location VUmc, Amsterdam Neuroscience, Amsterdam, the Netherlands.,Queen's Square Institute of Neurology and Centre for Medical Image Computing, University College London, London, UK
| | - Josef Pfeuffer
- Application Development, Siemens Healthcare, Erlangen, Germany
| | - Linda Knutsson
- Department of Medical Radiation Physics, Lund University, Lund, Sweden.,The Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Danielle van Westen
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden.,Diagnostic Radiology, Lund University, Lund, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden.,Department of Neurology, Skåne University Hospital, Lund, Sweden
| | - Henk Jmm Mutsaerts
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Location VUmc, Amsterdam Neuroscience, Amsterdam, the Netherlands.,Queen's Square Institute of Neurology and Centre for Medical Image Computing, University College London, London, UK
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
9
|
Hu Y, Zhang M, Liu B, Tang Y, Wang Z, Wang T, Zheng J, Zhang J. Honokiol prevents chronic cerebral hypoperfusion induced astrocyte A1 polarization to alleviate neurotoxicity by targeting SIRT3-STAT3 axis. Free Radic Biol Med 2023; 202:62-75. [PMID: 36997099 DOI: 10.1016/j.freeradbiomed.2023.03.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 03/11/2023] [Accepted: 03/20/2023] [Indexed: 04/01/2023]
Abstract
Alzheimer's Dementia (AD) and Vascular Dementia (VaD) are two main types of dementias for which no specific treatment is available. Chronic Cerebral Hypoperfusion (CCH) is a pathogenesis underlying AD and VaD that promotes neuroinflammatory responses and oxidative stress. Honokiol (HNK) is a natural compound isolated from magnolia leaves that can easily cross blood brain barrier and has anti-inflammatory and antioxidant effects. In the present study, the effects of HNK on astrocyte polarization and neurological damage in in vivo and in vitro models of chronic cerebral hypoperfusion were explored. We found that HNK was able to inhibit the phosphorylation and nuclear translocation of STAT3, A1 polarization, and reduce conditioned medium's neuronal toxicity of astrocyte under chronic hypoxia induced by cobalt chloride; STAT3 phosphorylation inhibitor C188-9 was able to mimic the above effects of HNK, suggesting that HNK may inhibit chronic hypoxia-induced A1 polarization in astrocytes via STAT3. SIRT3 inhibitor 3-TYP reversed, while Sirt3 overexpression mimicked the inhibitory effects of HNK on oxidative stress, STAT3 phosphorylation and nuclear translocation, A1 polarization and neuronal toxicity of astrocyte under chronic hypoxic conditions. For in vivo research, continuous intraperitoneal injection of HNK (1mg/kg) for 21 days ameliorated the decrease in SIRT3 activity and oxidative stress, inhibited astrocytic STAT3 nuclear translocation and A1 polarization, and prevented neuron and synaptic loss in the hippocampal of CCH rats. Besides, HNK application improved the spatial memory impairment of CCH rats, as assessed with Morris Water Maze. In conclusion, these results suggest that the phytochemical HNK can inhibit astrocyte A1 polarization via regulating SIRT3-STAT3 axis, thus improving CCH-induced neurological damage. These results highlight HNK as novel treatment for dementia with underlying vascular mechanisms.
Collapse
Affiliation(s)
- Yuan Hu
- Department of Neurology, Zhongnan Hospital of Wuhan University, Donghu Road No. 169, Wuhan, 430071, China.
| | - Miao Zhang
- Department of Neurology, Zhongnan Hospital of Wuhan University, Donghu Road No. 169, Wuhan, 430071, China
| | - Bihan Liu
- Department of Neurology, Zhongnan Hospital of Wuhan University, Donghu Road No. 169, Wuhan, 430071, China
| | - Yingying Tang
- Department of Neurology, Zhongnan Hospital of Wuhan University, Donghu Road No. 169, Wuhan, 430071, China
| | - Zhuo Wang
- Department of Neurology, Zhongnan Hospital of Wuhan University, Donghu Road No. 169, Wuhan, 430071, China
| | - Tao Wang
- Department of Neurology, First Clinical Medical College of China Three Gorges University, Yichang, Hubei, 443003, China
| | - Jiaxin Zheng
- Department of Neurology, Zhongnan Hospital of Wuhan University, Donghu Road No. 169, Wuhan, 430071, China
| | - Junjian Zhang
- Department of Neurology, Zhongnan Hospital of Wuhan University, Donghu Road No. 169, Wuhan, 430071, China.
| |
Collapse
|
10
|
Matsumoto H, Tagai K, Endo H, Matsuoka K, Takado Y, Kokubo N, Shimada H, Goto T, Goto TK, Higuchi M. Association of Tooth Loss with Alzheimer's Disease Tau Pathologies Assessed by Positron Emission Tomography. J Alzheimers Dis 2023; 96:1253-1265. [PMID: 37980663 PMCID: PMC10741329 DOI: 10.3233/jad-230581] [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] [Accepted: 09/24/2023] [Indexed: 11/21/2023]
Abstract
BACKGROUND Deterioration of the oral environment is one of the risk factors for dementia. A previous study of an Alzheimer's disease (AD) model mouse suggests that tooth loss induces denervation of the mesencephalic trigeminal nucleus and neuroinflammation, possibly leading to accelerated tau dissemination from the nearby locus coeruleus (LC). OBJECTIVE To elucidate the relevance of oral conditions and amyloid-β (Aβ) and tau pathologies in human participants. METHODS We examined the number of remaining teeth and the biofilm-gingival interface index in 24 AD-spectrum patients and 19 age-matched healthy controls (HCs). They also underwent positron emission tomography (PET) imaging of Aβ and tau with specific radiotracers, 11C-PiB and 18F-PM-PBB3, respectively. All AD-spectrum patients were Aβ-positive, and all HCs were Aβ-negative. We analyzed the correlation between the oral parameters and radiotracer retention. RESULTS No differences were found in oral conditions between the AD and HC groups. 11C-PiB retentions did not correlate with the oral indices in either group. In AD-spectrum patients, brain-wide, voxel-based image analysis highlighted several regions, including the LC and associated brainstem substructures, as areas where 18F-PM-PBB3 retentions negatively correlated with the remaining teeth and revealed the correlation of tau deposits in the LC (r = -0.479, p = 0.018) primarily with the hippocampal and neighboring areas. The tau deposition in none of the brain regions was associated with the periodontal status. CONCLUSIONS Our findings with previous preclinical evidence imply that tooth loss may enhance AD tau pathogenesis, promoting tau spreading from LC to the hippocampal formation.
Collapse
Affiliation(s)
- Hideki Matsumoto
- Department of Oral and Maxillofacial Radiology, Tokyo Dental College, Tokyo, Japan
- Department of Functional Brain Imaging Research, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Kenji Tagai
- Department of Functional Brain Imaging Research, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba, Japan
- Department of Psychiatry, The Jikei University of Medicine, Tokyo, Japan
| | - Hironobu Endo
- Department of Functional Brain Imaging Research, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Kiwamu Matsuoka
- Department of Functional Brain Imaging Research, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Yuhei Takado
- Department of Functional Brain Imaging Research, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Naomi Kokubo
- Department of Functional Brain Imaging Research, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Hitoshi Shimada
- Department of Functional Brain Imaging Research, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba, Japan
- Department of Functional Neurology & Neurosurgery, Center for Integrated Human Brain Science, Brain Research Institute, Niigata University, Niigata, Japan
| | - Tetsuya Goto
- Department of Oral Anatomy and Cell Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Tazuko K. Goto
- Department of Oral and Maxillofacial Radiology, Tokyo Dental College, Tokyo, Japan
- Department of Functional Brain Imaging Research, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba, Japan
- Tokyo Dental College Research Branding Project, Tokyo Dental College, Tokyo, Japan
- Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Makoto Higuchi
- Department of Functional Brain Imaging Research, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba, Japan
| |
Collapse
|
11
|
Su SH, Wu YF, Lin Q, Zhang L, Wang DP, Hai J. Fecal microbiota transplantation and replenishment of short-chain fatty acids protect against chronic cerebral hypoperfusion-induced colonic dysfunction by regulating gut microbiota, differentiation of Th17 cells, and mitochondrial energy metabolism. J Neuroinflammation 2022; 19:313. [PMID: 36567333 PMCID: PMC9791754 DOI: 10.1186/s12974-022-02675-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 12/17/2022] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Little is known about the association between gut microbiota and intestinal injury under a state of chronic cerebral hypoperfusion (CCH). Here, the effects of gut microbiota and short-chain fatty acids (SCFAs), as important metabolic products, on intestinal function and potential mechanisms after CCH were investigated. METHODS Rats were subjected to bilateral common carotid artery occlusion (BCCAo) to induce CCH. The gut microbiota and metabolites of SCFAs were assessed by 16S rRNA sequencing and targeted metabolomics, respectively. Transcriptomic analysis of colon tissues was also conducted. Subsequently, potential molecular pathways and differentially expressed genes were verified by western blot, immunoprecipitation, and immunofluorescence analyses. Furthermore, the integrity of the colonic barrier was evaluated by hematoxylin and eosin and mucin 2 staining and expression levels of tight junction proteins. Besides, colonic inflammation was further assessed by flow cytometry and expression levels of inflammatory cytokines. In addition, colonic mitochondrial dysfunction was analyzed via membrane potential, reactive oxygen species, electron transport chain (ETC) activities, adenosine triphosphate content, and mitochondrial ultrastructure. RESULTS CCH modified gut microbial composition and microbial metabolism of SCFAs, which may be associated with inhibition of mitochondrial ETC activities and oxidative phosphorylation, leading to dysregulation of mitochondrial energy metabolism. Furthermore, CCH induced differentiation of pathogenic Th17 cells, promoted the formation of complexes of interferon regulatory factor 4 and signal transducer and activator of transcription 3 (STAT3), and increased the phosphorylation of STAT3. This was associated with an impairment of colonic barrier function and chronic colonic inflammation. In contrast, FMT and SCFA replenishment ameliorated CCH-induced gut microbial dysbiosis by increasing the intestinal content of Ruminococcus_sp_N15_MGS_57 and modulating microbial metabolism of SCFAs by increasing acetic acid contents associated with an improvment of the balance between Tregs and Th17 cells, mitochondrial ETC activities, and oxidative phosphorylation to prevent colonic inflammation and dysregulation of mitochondrial energy metabolism. CONCLUSION These findings indicate that FMT and SCFA replenishment present a promising therapeutic strategy against colonic dysfunction under a state of chronic cerebral ischemia.
Collapse
Affiliation(s)
- Shao-Hua Su
- grid.24516.340000000123704535Department of Neurosurgery, School of Medicine, Tongji Hospital, Tongji University, 389 Xincun Road, Shanghai, 200065 China
| | - Yi-Fang Wu
- grid.24516.340000000123704535Department of Neurosurgery, School of Medicine, Tongji Hospital, Tongji University, 389 Xincun Road, Shanghai, 200065 China
| | - Qi Lin
- grid.16821.3c0000 0004 0368 8293Department of Pharmacy, School of Medicine, Institutes of Medical Sciences, Shanghai Jiao Tong University, Shanghai, 200025 China
| | - Lin Zhang
- grid.16821.3c0000 0004 0368 8293Department of Neurosurgery, School of Medicine, Shanghai Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai, 200003 China
| | - Da-Peng Wang
- grid.24516.340000000123704535Department of Neurosurgery, School of Medicine, Tongji Hospital, Tongji University, 389 Xincun Road, Shanghai, 200065 China
| | - Jian Hai
- grid.24516.340000000123704535Department of Neurosurgery, School of Medicine, Tongji Hospital, Tongji University, 389 Xincun Road, Shanghai, 200065 China
| |
Collapse
|
12
|
Arias JC, Edwards M, Vitali F, Beach TG, Serrano GE, Weinkauf CC. Extracranial carotid atherosclerosis is associated with increased neurofibrillary tangle accumulation. J Vasc Surg 2022; 75:223-228. [PMID: 34478810 PMCID: PMC8976507 DOI: 10.1016/j.jvs.2021.07.238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 07/25/2021] [Indexed: 01/03/2023]
Abstract
OBJECTIVE We sought to determine whether extracranial carotid atherosclerotic disease (ECAD) is associated with increased key neurodegenerative pathology such as neurofibrillary tangle (NFT), beta-amyloid plaque, or cerebral amyloid angiopathy (CAA) accumulation, findings associated with Alzheimer's disease (AD) and other dementias. METHODS Our prospective, longitudinal, clinicopathologic study, the AZSAND (Arizona study of aging and neurodegenerative disorders) and Brain and Body Donation Program, recorded the presence or absence of clinically diagnosed ECAD and performed semiquantitative density estimates of NFT, beta-amyloid plaque, and CAA at death. After adjusting for potential confounding factors determined by logistic regression analysis, histopathology density scores were evaluated in individuals with ECAD (n = 66) and those without ECAD (n = 125). RESULTS We found that the presence of ECAD was associated with a 21% greater NFT burden at death compared with no ECAD (P = .02). Anatomically, an increased NFT burden was seen throughout the brain regions evaluated but was significant in the temporal lobe (P < .05) and entorhinal cortex (P = .02). In addition, we found that subjects who had undergone carotid endarterectomy (CEA), the surgical treatment of ECAD (n = 32), had decreased NFT densities compared with those with ECAD who had not undergone CEA (n = 66; P = .04). In contrast to NFT, ECAD was not associated with beta-amyloid plaques or CAA density. CONCLUSIONS These findings indicate that ECAD is associated with the NFT burden in the temporal lobe and entorhinal cortex, which has clinical significance for AD and non-AD dementias and cognitive dysfunction. Further understanding of whether ECAD increases the risk of neurodegenerative brain changes is highly relevant because ECAD is a treatable disease that has not, otherwise, been evaluated for nor specifically treated as a dementia risk factor.
Collapse
Affiliation(s)
- Juan C. Arias
- Department of Surgery, University of Arizona, Tucson, Arizona, USA
| | - Mark Edwards
- Department of Surgery, University of Arizona, Tucson, Arizona, USA
| | - Francesca Vitali
- Center for Innovation in Brain Science; University of Arizona, Tucson, Arizona, USA.,Department of Neurology; University of Arizona College of Medicine, Tucson, Arizona, USA.,Center for Biomedical Informatics and Biostatistics; University of Arizona, Tucson, Arizona, USA
| | - Thomas G. Beach
- Banner Sun Health Research Institute, Sun City, Arizona, USA
| | | | | |
Collapse
|
13
|
Scheffer S, Hermkens DMA, van der Weerd L, de Vries HE, Daemen MJAP. Vascular Hypothesis of Alzheimer Disease: Topical Review of Mouse Models. Arterioscler Thromb Vasc Biol 2021; 41:1265-1283. [PMID: 33626911 DOI: 10.1161/atvbaha.120.311911] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Sanny Scheffer
- Department of Pathology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, the Netherlands (S.S., D.M.A.H., M.J.A.P.D.)
| | - Dorien M A Hermkens
- Department of Pathology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, the Netherlands (S.S., D.M.A.H., M.J.A.P.D.)
| | - Louise van der Weerd
- Departments of Radiology & Human Genetics, Leiden University Medical Center, the Netherlands (L.v.d.W.)
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, Amsterdam UMC, Vrije University of Amsterdam, the Netherlands (H.E.d.V.)
| | - Mat J A P Daemen
- Department of Pathology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, the Netherlands (S.S., D.M.A.H., M.J.A.P.D.)
| |
Collapse
|
14
|
de la Torre JC. Deciphering Alzheimer's Disease Pathogenic Pathway: Role of Chronic Brain Hypoperfusion on p-Tau and mTOR. J Alzheimers Dis 2021; 79:1381-1396. [PMID: 33459641 DOI: 10.3233/jad-201165] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
This review examines new biomolecular findings that lend support to the hemodynamic role played by chronic brain hypoperfusion (CBH) in driving a pathway to Alzheimer's disease (AD). CBH is a common clinical feature of AD and the current topic of intense investigation in AD models. CBH is also the basis for the vascular hypothesis of AD which we originally proposed in 1993. New biomolecular findings reveal the interplay of CBH in increasing tau phosphorylation (p-Tau) in the hippocampus and cortex of AD mice, damaging fast axonal transport, increasing signaling of mammalian target of rapamycin (mTOR), impairing learning-memory function, and promoting the formation of neurofibrillary tangles, a neuropathologic hallmark of AD. These pathologic elements have been singularly linked with neurodegeneration and AD but their abnormal, collective participation during brain aging have not been fully examined. The format for this review will provide a consolidated analysis of each pathologic phase contributing to cognitive decline and AD onset, summarized in nine chronological steps. These steps galvanize each factor's active participation and contribution in constructing a biomolecular pathway to AD onset generated by CBH.
Collapse
Affiliation(s)
- Jack C de la Torre
- Department of Psychology, University of Texas at Austin, Austin, TX, USA.,Department of Physiology, University of Valencia Faculty of Medicine, Valencia, Spain
| |
Collapse
|
15
|
Albrecht D, Isenberg AL, Stradford J, Monreal T, Sagare A, Pachicano M, Sweeney M, Toga A, Zlokovic B, Chui H, Joe E, Schneider L, Conti P, Jann K, Pa J. Associations between Vascular Function and Tau PET Are Associated with Global Cognition and Amyloid. J Neurosci 2020; 40:8573-8586. [PMID: 33046556 PMCID: PMC7605425 DOI: 10.1523/jneurosci.1230-20.2020] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 08/10/2020] [Accepted: 08/19/2020] [Indexed: 11/21/2022] Open
Abstract
Tau pathology and vascular dysfunction are important contributors to Alzheimer's disease (AD), but vascular-tau associations and their effects on cognition are poorly understood. We investigated these associations in male and female humans by conducting voxelwise comparisons between cerebral blood flow (CBF) and tau positron emission tomography (PET) images in independent discovery [cognitively normal (CN), 19; mild cognitive impairment (MCI) risk, 43; MCI, 6] and replication (CN,73; MCI, 45; AD, 20) cohorts. In a subgroup, we assessed relationships between tau and soluble platelet-derived growth factor β (sPDGFRβ), a CSF marker of pericyte injury. We tested whether CBF/sPDGFRβ-tau relationships differed based on Montreal Cognitive Assessment (MoCA) global cognition performance, or based on amyloid burden. Mediation analyses assessed relationships among CBF/sPDGFRβ, tau, and cognition. Negative CBF-tau correlations were observed predominantly in temporal-parietal regions. In the replication cohort, early negative CBF-tau correlations increased in spatial extent and in strength of correlation with increased disease severity. Stronger CBF-tau and sPDGFRβ-tau correlations were observed in participants with greater amyloid burden and lower MoCA scores. Importantly, when stratifying by amyloid status, stronger CBF-tau relationships in individuals with lower MoCA scores were driven by amyloid+ participants. Tau PET was a significant mediator CBF/sPDGFRβ-MoCA relationships in numerous regions. Our results demonstrate vascular-tau associations across the AD spectrum and suggest that early vascular-tau associations are exacerbated in the presence of amyloid, consistent with a two-hit model of AD on cognition. Combination treatments targeting vascular health, as well as amyloid-β and tau levels, may preserve cognitive function more effectively than single-target therapies.SIGNIFICANCE STATEMENT Emerging evidence demonstrates a role for vascular dysfunction as a significant contributor to Alzheimer's pathophysiology. However, associations between vascular dysfunction and tau pathology, and their effects on cognition remain poorly understood. Multimodal neuroimaging data from two independent cohorts were analyzed to provide novel in vivo evidence of associations between cerebral blood flow (CBF), an MRI measure of vascular health, and tau pathology using PET. CBF-tau associations were related to cognition and driven in part by amyloid burden. Soluble platelet-derived growth factor β, an independent CSF vascular biomarker, confirmed vascular-tau associations in a subgroup analysis. These results suggest that combination treatments targeting vascular health, amyloid-β, and tau levels may more effectively preserve cognitive function than single-target therapies.
Collapse
Affiliation(s)
- Daniel Albrecht
- Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, California 90033
| | - A Lisette Isenberg
- Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, California 90033
| | - Joy Stradford
- Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, California 90033
| | - Teresa Monreal
- Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, California 90033
| | - Abhay Sagare
- Department of Physiology and Neuroscience and the Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California 90033
| | - Maricarmen Pachicano
- Department of Physiology and Neuroscience and the Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California 90033
| | - Melanie Sweeney
- Department of Physiology and Neuroscience and the Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California 90033
| | - Arthur Toga
- Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, California 90033
| | - Berislav Zlokovic
- Department of Physiology and Neuroscience and the Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California 90033
| | - Helena Chui
- Alzheimer Disease Research Center, Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, California 90033
| | - Elizabeth Joe
- Alzheimer Disease Research Center, Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, California 90033
| | - Lon Schneider
- Alzheimer Disease Research Center, Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, California 90033
| | - Peter Conti
- Molecular Imaging Center, Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, California 90033
| | - Kay Jann
- Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, California 90033
| | - Judy Pa
- Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, California 90033
- Alzheimer Disease Research Center, Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, California 90033
| |
Collapse
|
16
|
Shindo A, Ishikawa H, Ii Y, Niwa A, Tomimoto H. Clinical Features and Experimental Models of Cerebral Small Vessel Disease. Front Aging Neurosci 2020; 12:109. [PMID: 32431603 PMCID: PMC7214616 DOI: 10.3389/fnagi.2020.00109] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 03/30/2020] [Indexed: 12/24/2022] Open
Abstract
Cerebral small vessel disease (SVD) refers to a group of disease conditions affecting the cerebral small vessels, which include the small arteries, arterioles, capillaries, and postcapillary venules in the brain. SVD is the primary cause of vascular cognitive impairment and gait disturbances in aged people. There are several types of SVD, though arteriolosclerosis, which is mainly associated with hypertension, aging, and diabetes mellitus, and cerebral amyloid angiopathy (CAA) comprise most SVD cases. The pathology of arteriolosclerosis-induced SVD is characterized by fibrinoid necrosis and lipohyalinosis, while CAA-associated SVD is characterized by progressive deposition of amyloid beta (Aβ) protein in the cerebral vessels. Brain magnetic resonance imaging (MRI) has been used for examination of SVD lesions; typical lesions are characterized by white matter hyperintensity, lacunar infarcts, enlargement of perivascular spaces (EPVS), microbleeds, cortical superficial siderosis (cSS), and cortical microinfarcts. The microvascular changes that occur in the small vessels are difficult to identify clearly; however, these consequent image findings can represent the SVD. There are two main strategies for prevention and treatment of SVD, i.e., pharmacotherapy and lifestyle modification. In this review, we discuss clinical features of SVD, experimental models replicating SVD, and treatments to further understand the pathological and clinical features of SVD.
Collapse
Affiliation(s)
- Akihiro Shindo
- Department of Neurology, Mie University Graduate School of Medicine, Mie University, Tsu, Japan
| | - Hidehiro Ishikawa
- Department of Neurology, Mie University Graduate School of Medicine, Mie University, Tsu, Japan
| | - Yuichiro Ii
- Department of Neurology, Mie University Graduate School of Medicine, Mie University, Tsu, Japan
| | - Atsushi Niwa
- Department of Neurology, Mie University Graduate School of Medicine, Mie University, Tsu, Japan
| | - Hidekazu Tomimoto
- Department of Neurology, Mie University Graduate School of Medicine, Mie University, Tsu, Japan
| |
Collapse
|
17
|
Cheng Y, Buchan M, Vitanova K, Aitken L, Gunn-Moore FJ, Ramsay RR, Doherty G. Neuroprotective actions of leptin facilitated through balancing mitochondrial morphology and improving mitochondrial function. J Neurochem 2020; 155:191-206. [PMID: 32157699 DOI: 10.1111/jnc.15003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 03/02/2020] [Accepted: 03/03/2020] [Indexed: 12/12/2022]
Abstract
Mitochondrial dysfunction has a recognised role in the progression of Alzheimer's disease (AD) pathophysiology. Cerebral perfusion becomes increasingly inefficient throughout ageing, leading to unbalanced mitochondrial dynamics. This effect is exaggerated by amyloid β (Aβ) and phosphorylated tau, two hallmark proteins of AD pathology. A neuroprotective role for the adipose-derived hormone, leptin, has been demonstrated in neuronal cells. However, its effects with relation to mitochondrial function in AD remain largely unknown. To address this question, we have used both a glucose-serum-deprived (CGSD) model of ischaemic stroke in SH-SY5Y cells and a Aβ1-42 -treatment model of AD in differentiated hippocampal cells. Using a combination of 5,5',6,6'-tetrachloro-1,1',3,3'-tetraethylbenzimidazolylcarbocyanine iodide (JC-1) and MitoRed staining techniques, we show that leptin prevents depolarisation of the mitochondrial membrane and excessive mitochondrial fragmentation induced by both CGSD and Aβ1-42 . Thereafter, we used ELISAs and a number of activity assays to reveal the biochemical underpinnings of these processes. Specifically, leptin was seen to inhibit up-regulation of the mitochondrial fission protein Fis1 and down-regulation of the mitochondrial fusion protein, Mfn2. Furthermore, leptin was seen to up-regulate the expression and activity of the antioxidant enzyme, monoamine oxidase B. Herein we provide the first demonstration that leptin is sufficient to protect against aberrant mitochondrial dynamics and resulting loss of function induced by both CGSD and Aβ1-42 . We conclude that the established neuroprotective actions of leptin may be facilitated through regulation of mitochondrial dynamics.
Collapse
Affiliation(s)
- Ying Cheng
- School of Psychology and Neuroscience, University of St Andrews, St Andrews, UK
| | - Matthew Buchan
- School of Psychology and Neuroscience, University of St Andrews, St Andrews, UK
| | - Karina Vitanova
- School of Psychology and Neuroscience, University of St Andrews, St Andrews, UK
| | - Laura Aitken
- School of Biology, University of St Andrews, St Andrews, UK
| | | | - Rona R Ramsay
- School of Biology, University of St Andrews, St Andrews, UK
| | - Gayle Doherty
- School of Psychology and Neuroscience, University of St Andrews, St Andrews, UK
| |
Collapse
|