1
|
Schroevers JL, Eggink E, Hoevenaar-Blom MP, Van Dalen JW, Van Middelaar T, Van Gool WA, Richard E, Moll Van Charante EP. Antihypertensive medication classes and the risk of dementia over a decade of follow-up. J Hypertens 2023; 41:262-270. [PMID: 36394298 PMCID: PMC9799049 DOI: 10.1097/hjh.0000000000003324] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/27/2022] [Accepted: 10/18/2022] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Use of angiotensin II (ATII)-stimulating antihypertensive medication (AHM), including angiotensin receptor blockers (ARBs) and dihydropyridine calcium channel blockers (CCBs), has been associated with lower dementia risk. Previous studies had relatively short follow-up periods. The aim of this study is to investigate if these effects are sustained over longer periods. METHODS This post hoc observational analysis was based on data from a dementia prevention trial (preDIVA and its observational extension), among Dutch community-dwelling older adults without prior diagnosis of dementia. Differential associations between AHM classes and incident dementia were studied after 7.0 and 10.4 years, based on the median follow-up durations of dementia cases and all participants. RESULTS After 7 years, use of ATII-stimulating antihypertensives [hazard ratio = 0.68, 95% confidence interval (CI) = 0.47-1.00], ARBs (hazard ratio = 0.54, 95% CI = 0.31-0.94) and dihydropyridine CCBs (hazard ratio = 0.52, 95% CI = 0.30-0.91) was associated with lower dementia risk. After 10.4 years, associations for ATII-stimulating antihypertensives, ARBs and dihydropyridine CCBs attenuated (hazard ratio = 0.80, 95% CI = 0.61-1.04; hazard ratio = 0.75, 95% CI = 0.53-1.07; hazard ratio = 0.73, 95% CI = 0.51-1.04 respectively), but still suggested lower dementia risk when compared with use of other AHM classes. Results could not be explained by competing risk of mortality. CONCLUSION Our results suggest that use of ARBs, dihydropyridine CCBs and ATII-stimulating antihypertensives is associated with lower dementia risk over a decade, although associations attenuate over time. Apart from methodological aspects, differential effects of antihypertensive medication classes on incident dementia may in part be temporary, or decrease with ageing.
Collapse
Affiliation(s)
| | - Esmé Eggink
- Department of General Practice/Family Medicine
| | - Marieke P. Hoevenaar-Blom
- Department of Public and Occupational Health, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam
| | - Jan Willem Van Dalen
- Department of Neurology, Donders Institute for Brain, Behaviour and Cognition, Radboud University Medical Centre, Nijmegen
| | - Tessa Van Middelaar
- Department of Neurology, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, the Netherlands
| | - Willem A. Van Gool
- Department of Public and Occupational Health, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam
| | - Edo Richard
- Department of Public and Occupational Health, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam
- Department of Neurology, Donders Institute for Brain, Behaviour and Cognition, Radboud University Medical Centre, Nijmegen
| | - Eric P. Moll Van Charante
- Department of General Practice/Family Medicine
- Department of Public and Occupational Health, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam
| |
Collapse
|
2
|
Xu J, Huang X, Guo Y, Ma X, Li P, Zhou S, Zhang C, Chen R, Van Halm-Lutterodt N, Yuan L. Discrepant modulating effects of dietary docosahexaenoic acid on cerebral lipids, fatty acid transporter expression and soluble beta-amyloid levels in ApoE -/- and C57BL/6J mice. Neuropathol Appl Neurobiol 2023; 49:e12855. [PMID: 36259948 DOI: 10.1111/nan.12855] [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: 04/01/2022] [Revised: 08/03/2022] [Accepted: 08/21/2022] [Indexed: 11/30/2022]
Abstract
AIMS The study was designed to explore the role of apolipoprotein E (ApoE) deficiency concomitant with dietary docosahexaenoic acid (DHA) treatment on brain β-amyloid (Aβ) and lipid levels. METHOD A 5-month dietary DHA intervention was conducted in ApoE-deficient (ApoE-/- ) mice and wild-type C57BL/6J (C57 wt) mice. The Morris water maze test was performed to assess the behaviour of the animals. The cortical contents of soluble Aβ1-40 and Aβ1-42 were detected by enzyme-linked immunosorbent assay (ELISA). Cortical fatty acid levels were detected by gas chromatography. Gene and protein expression of molecules associated with cerebral Aβ and lipid metabolism were measured using real-time polymerase chain reaction (PCR), Western blot and histological methods. RESULTS DHA treatment increased the content of cortical DHA and n-3 polyunsaturated fatty acids (n-3 PUFAs) but decreased the ratio of n-6/n-3 PUFAs in ApoE-/- mice; whereas the content of cortical DHA and n-3 PUFAs in C57 wt mice remained unchanged after DHA treatment. Cerebral Fabp5 and Cd36 gene expression were significantly downregulated in DHA-fed C57 wt mice; cerebral Cd36 and Scarb1 gene expression were significantly upregulated, whereas Fabp5 gene expression was downregulated in DHA-fed ApoE-/- mice. In comparison with C57 wt mice, the content of cortical soluble Aβ1-42 , total cholesterol (TC) and low-density lipoprotein cholesterol (LDL-C) increased, whereas the level of high-density lipoprotein cholesterol (HDL-C) decreased in ApoE-/- mice. Interestingly, these differences were significantly reversed by DHA dietary treatment. CONCLUSION DHA intervention has discrepant impacts on cerebral lipids, fatty acid transporter expression and soluble Aβ levels in ApoE-/- and C57 wt mice, suggesting the modifying role of ApoE status on the responses of cerebral lipids and Aβ metabolism to DHA treatment.
Collapse
Affiliation(s)
- Jingjing Xu
- School of Public Health, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, China
| | - Xiaochen Huang
- School of Public Health, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, China.,Nutrition Department, Beijing Jishuitan Hospital, Beijing, China
| | - Yujie Guo
- School of Public Health, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, China
| | - Xiaojun Ma
- School of Public Health, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, China
| | - Pengfei Li
- School of Public Health, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, China
| | - Shaobo Zhou
- School of Science, Faculty of Engineering and Science, University of Greenwich, Chatham, UK
| | - Chi Zhang
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Rui Chen
- School of Public Health, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, China
| | | | - Linhong Yuan
- School of Public Health, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, China
| |
Collapse
|
3
|
Wharton W, Anderson A, Hayden KM, Carmichael OT, Clark JM, Luchsinger JA, Espeland M, Yasar S. Effect of renin-angiotensin system antihypertensive medication use on cognitive function in diabetes mellitus with obesity or overweight: An ancillary study to the Action for Health in Diabetes (Look AHEAD) trial. Diabetes Obes Metab 2022; 24:2443-2453. [PMID: 36065050 PMCID: PMC9617758 DOI: 10.1111/dom.14838] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 08/05/2022] [Accepted: 08/09/2022] [Indexed: 11/30/2022]
Abstract
AIM To determine whether antihypertensive medication (AHM) acting through the renin angiotensin system (RAS-AHM), compared with other AHM, can mitigate effects on cognitive function and risk for impairment in a population with type 2 diabetes mellitus (T2DM). MATERIALS AND METHODS This secondary analysis of the randomized controlled Action for Health in Diabetes (Look AHEAD) study included 712 community-dwelling participants who were followed over 15 years. Logistic regression was used to relate RAS-AHM use to cognitive impairment, and linear regression was used to relate RAS-AHM use to domain-specific cognitive function after adjusting for potential confounders. RESULTS A total of 563 individuals reported RAS-AHM use and 149 reported other-AHM use during the study. RAS-AHM users have college or higher education (53%), had higher baseline glycated haemoglobin (57 mmol/mol), and reported higher diabetes medication use (86%), while other-AHM users were more likely to be White (72%), obese (25%) and to have cardiovascular history (19%). RAS-AHM use was not associated with a reduced risk of dementia compared with other-AHM use. We did observe better executive function (Trail Making Test, part B, P < 0.04), processing speed (Digit Symbol Substitution Test, P < 0.004), verbal memory (Rey Auditory Verbal Learning Test-delayed recall, P < 0.005), and composite score (P < 0.008) among RAS-AHM users compared with other-AHM users. CONCLUSION In this sample of adults with T2DM, free of dementia at baseline, we observed a slower decline in processing speed, executive function, verbal memory, and composite score among RAS-AHM users.
Collapse
Affiliation(s)
| | - Andrea Anderson
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Kathleen M. Hayden
- Department of Social Sciences and Health Policy, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Owen T Carmichael
- Biomedical Imaging Center, Pennington Biomedical Research Center, Baton Rouge, LA
| | - Jeanne M Clark
- Division of General Internal Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - José A. Luchsinger
- Departments of Medicine and Epidemiology, Columbia University Irving Medical Center, New York City, NY, USA
| | - Mark Espeland
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Section of Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Sevil Yasar
- Division of Gerontology and Geriatric Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| |
Collapse
|
4
|
Meng X, Wei Q, Meng L, Liu J, Wu Y, Liu W. Feature Fusion and Detection in Alzheimer's Disease Using a Novel Genetic Multi-Kernel SVM Based on MRI Imaging and Gene Data. Genes (Basel) 2022; 13:837. [PMID: 35627222 PMCID: PMC9140721 DOI: 10.3390/genes13050837] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 05/03/2022] [Accepted: 05/05/2022] [Indexed: 01/27/2023] Open
Abstract
Voxel-based morphometry provides an opportunity to study Alzheimer's disease (AD) at a subtle level. Therefore, identifying the important brain voxels that can classify AD, early mild cognitive impairment (EMCI) and healthy control (HC) and studying the role of these voxels in AD will be crucial to improve our understanding of the neurobiological mechanism of AD. Combining magnetic resonance imaging (MRI) imaging and gene information, we proposed a novel feature construction method and a novel genetic multi-kernel support vector machine (SVM) method to mine important features for AD detection. Specifically, to amplify the differences among AD, EMCI and HC groups, we used the eigenvalues of the top 24 Single Nucleotide Polymorphisms (SNPs) in a p-value matrix of 24 genes associated with AD for feature construction. Furthermore, a genetic multi-kernel SVM was established with the resulting features. The genetic algorithm was used to detect the optimal weights of 3 kernels and the multi-kernel SVM was used after training to explore the significant features. By analyzing the significance of the features, we identified some brain regions affected by AD, such as the right superior frontal gyrus, right inferior temporal gyrus and right superior temporal gyrus. The findings proved the good performance and generalization of the proposed model. Particularly, significant susceptibility genes associated with AD were identified, such as CSMD1, RBFOX1, PTPRD, CDH13 and WWOX. Some significant pathways were further explored, such as the calcium signaling pathway (corrected p-value = 1.35 × 10-6) and cell adhesion molecules (corrected p-value = 5.44 × 10-4). The findings offer new candidate abnormal brain features and demonstrate the contribution of these features to AD.
Collapse
Affiliation(s)
- Xianglian Meng
- School of Computer Information and Engineering, Changzhou Institute of Technology, Changzhou 213032, China; (X.M.); (Q.W.); (J.L.); (Y.W.)
| | - Qingpeng Wei
- School of Computer Information and Engineering, Changzhou Institute of Technology, Changzhou 213032, China; (X.M.); (Q.W.); (J.L.); (Y.W.)
| | - Li Meng
- School of Physics, Engineering and Computer Science, University of Hertfordshire, Hatfield AL10 9AB, UK
| | - Junlong Liu
- School of Computer Information and Engineering, Changzhou Institute of Technology, Changzhou 213032, China; (X.M.); (Q.W.); (J.L.); (Y.W.)
| | - Yue Wu
- School of Computer Information and Engineering, Changzhou Institute of Technology, Changzhou 213032, China; (X.M.); (Q.W.); (J.L.); (Y.W.)
| | - Wenjie Liu
- School of Computer Information and Engineering, Changzhou Institute of Technology, Changzhou 213032, China; (X.M.); (Q.W.); (J.L.); (Y.W.)
| |
Collapse
|
5
|
Baron DH, Skrobot OA, Palmer JC, Sharma K, Kehoe P. The Renin Angiotensin System as a potential treatment target for Traumatic Brain Injury. J Neurotrauma 2022; 39:473-486. [PMID: 35029131 DOI: 10.1089/neu.2021.0401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022] Open
Abstract
Traumatic brain injury (TBI) is a major health concern and leading cause of death and disability in young adults in the UK and worldwide, however, there is a paucity of disease modifying therapies for the treatment of TBI. This review investigates the potential of the renin-angiotensin system (RAS) as a treatment pathway for traumatic brain injury (TBI) in adults. Relevant electronic databases were searched on 18 December 2019, updated 16 May 2021. All English language articles with adult human or animal populations investigating RAS drugs as an intervention for TBI or reporting genetic evidence relevant to the RAS and TBI were screened. Eighteen preclinical RCTs (n=2269) and 2 clinical cohort studies (n=771) were included. Meta-analyses of 6 preclinical randomised-controlled trials (n=99) indicated candesartan improved neurological function short-term (<7 days: standardised mean difference (SMD) 0.61, 95% confidence interval (CI) 0.19 - 1.03, I2=0%) and long-term (≥7 days: SMD 1.06, 95% CI 0.38; 1.73, I2=0%) post-TBI. Findings were similar for most secondary outcomes. There was a suggestion of benefit from other RAS-targeting drugs, although evidence was limited due to few small studies. There was insufficient evidence to enable strong assessment of these drugs on mortality post-TBI. We conclude that angiotensin-receptor blockers, especially candesartan, show positive outcomes post-TBI in preclinical studies with moderate quality of evidence (GRADE). More research into the effect of regulatory-RAS targeting drugs is needed. Clinical trials of candesartan following TBI are recommended, due to strong and consistent evidence of neuroprotection shown by these preclinical studies.
Collapse
Affiliation(s)
- Daniel Hendrik Baron
- University of Bristol, 1980, Dementia Research Group, Clinical Neurosciences, School of Clinical Sciences, Bristol, United Kingdom of Great Britain and Northern Ireland;
| | - Olivia A Skrobot
- University of Bristol, 1980, Translational Health Sciences, Dementia Research Group, Clinical Neurosciences, School of Clinical Sciences, Bristol, Bristol, United Kingdom of Great Britain and Northern Ireland;
| | - Jennifer C Palmer
- University of Bristol, 1980, Population Health Sciences, Bristol Medical School, Bristol, Bristol, United Kingdom of Great Britain and Northern Ireland.,University of Bristol, 1980, MRC Integrative Epidemiology Unit, Bristol, Bristol, United Kingdom of Great Britain and Northern Ireland;
| | - Kanchan Sharma
- University of Bristol, 1980, Translational Health Sciences, Bristol, Bristol, United Kingdom of Great Britain and Northern Ireland.,North Bristol NHS Trust, 1982, Neurology, Westbury on Trym, Bristol, United Kingdom of Great Britain and Northern Ireland;
| | - Patrick Kehoe
- University of Bristol, 1980, Translational Health Sciences, Dementia Research Group,, Clinical Neurosciences, School of Clinical Sciences, Bristol, - None -, United Kingdom of Great Britain and Northern Ireland, BS10 5NB.,University of Bristol;
| |
Collapse
|
6
|
Yasar S, Moored KD, Adam A, Zabel F, Chuang YF, Varma VR, Carlson MC. Angiotensin II Blood Levels Are Associated with Smaller Hippocampal and Cortical Volumes in Cognitively Normal Older Adults. J Alzheimers Dis 2021; 75:521-529. [PMID: 32280103 DOI: 10.3233/jad-200118] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
BACKGROUND There is emerging evidence about possible involvement of the renin-angiotensin system (RAS) in the pathogenesis of Alzheimer's disease (AD) and decline of cognitive function. However, little is known about associations with brain biomarkers. OBJECTIVE Our study aimed to examine associations between blood ACE-1 and ANG II levels and brain MRI based volumes in non-demented participants, and whether these associations were mediated by blood pressure. METHODS This cross-sectional study was conducted in 34 older participants from the Baltimore Experience Corps Trial (BECT) Brain Health Sub-study (BHS). Blood ANGII and ACE-1 levels were measured by ELISA and brain MRI volumes were generated using FreeSurfer 6.0. Multiple linear regression analysis, adjusting for intracranial volume and confounders, was used to determine associations between log transformed ANGII and ACE-1 levels and MRI volumes (mm3). RESULTS Participants were predominantly female (76%), African-American (94%), with mean age of 66.9 and education of 14.4 years. In the fully adjusted model we observed significant inverse associations between log ANGII levels and total grey matter (β=Angiotensin II associated with smaller hippocampus14,935.50, ±7,444.83, p = 0.05), total hippocampus (β=-129.97, ±105.27, p = 0.03), rostral middle frontal (β= -1580.40, ±584.74, p = 0.02), and supramarginal parietal (β= -978.90, ±365.54, p = 0.02) volumes. There were no associations between ANGII levels and total white matter or entorhinal cortex volumes, or ACE-1 levels and any brain volumes. CONCLUSION We observed that increased blood ANGII levels were associated with lower total grey matter, hippocampal, rostral middle frontal, and supramarginal parietal volumes, which are associated with cognitive domains that decline in preclinical AD.
Collapse
Affiliation(s)
- Sevil Yasar
- Department of Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Kyle D Moored
- Department of Mental Health, Johns Hopkins University, Bloomberg School of Public Health, Baltimore, MD, USA
| | - Atif Adam
- Department of Mental Health, Johns Hopkins University, Bloomberg School of Public Health, Baltimore, MD, USA
| | - Fiona Zabel
- Baltimore Polytechnic Institute, Baltimore, MD, USA
| | - Yi-Fang Chuang
- Institute of Public Health, National Yang-Ming University, Taipei, Taiwan
| | - Vijay R Varma
- National Institute on Aging, Intramural Research program, Laboratory of Behavioral Neuroscience, Baltimore, MD, USA
| | - Michelle C Carlson
- Department of Mental Health, Johns Hopkins University, Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
7
|
Engin AB, Engin ED, Engin A. Current opinion in neurological manifestations of SARS-CoV-2 infection. CURRENT OPINION IN TOXICOLOGY 2021; 25:49-56. [PMID: 33817451 PMCID: PMC8006515 DOI: 10.1016/j.cotox.2021.03.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Neurological symptoms occur in approximately one-third of hospitalized patients with coronavirus disease 2019 (COVID-19). Among these symptoms, hypoxic encephalopathy develops in one-fifth of severe cases, while ischemic strokes due to thrombotic complications are common in one-third of COVID-19 intensive care patients. Brain involvement of severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) is eventuated by several routes, including hematogenous spread, transsynaptic entry through infected neurons, olfactory nerve, ocular epithelium, vascular endothelium, and impaired blood–brain barrier. Besides the high angiotensin-converting enzyme-2 (ACE2) binding affinity, and FURIN preactivation, SARS-CoV-2 maintains efficient neuronal entry while evading immune surveillance by using basigin and neuropilin-1 receptors. However, the neurological manifestations and their pathogenic mechanisms are still debated in COVID-19 patients.
Collapse
Affiliation(s)
- Ayse Basak Engin
- Gazi University, Faculty of Pharmacy, Department of Toxicology, Ankara, Turkey
| | - Evren Doruk Engin
- Ankara University, Biotechnology Institute, Gumusdere Campus, Kecioren, Ankara, Turkey
| | - Atilla Engin
- Gazi University, Faculty of Medicine, Department of General Surgery, Ankara, Turkey
| |
Collapse
|
8
|
Calabrò M, Rinaldi C, Santoro G, Crisafulli C. The biological pathways of Alzheimer disease: a review. AIMS Neurosci 2020; 8:86-132. [PMID: 33490374 PMCID: PMC7815481 DOI: 10.3934/neuroscience.2021005] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 11/23/2020] [Indexed: 12/18/2022] Open
Abstract
Alzheimer disease is a progressive neurodegenerative disorder, mainly affecting older people, which severely impairs patients' quality of life. In the recent years, the number of affected individuals has seen a rapid increase. It is estimated that up to 107 million subjects will be affected by 2050 worldwide. Research in this area has revealed a lot about the biological and environmental underpinnings of Alzheimer, especially its correlation with β-Amyloid and Tau related mechanics; however, the precise molecular events and biological pathways behind the disease are yet to be discovered. In this review, we focus our attention on the biological mechanics that may lie behind Alzheimer development. In particular, we briefly describe the genetic elements and discuss about specific biological processes potentially associated with the disease.
Collapse
Affiliation(s)
| | | | | | - Concetta Crisafulli
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Italy
| |
Collapse
|
9
|
Wright JW, Harding JW. Contributions by the Brain Renin-Angiotensin System to Memory, Cognition, and Alzheimer's Disease. J Alzheimers Dis 2020; 67:469-480. [PMID: 30664507 DOI: 10.3233/jad-181035] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by progressive neuron losses in memory-associated brain structures that rob patients of their dignity and quality of life. Five drugs have been approved by the FDA to treat AD but none modify or significantly slow disease progression. New therapies are needed to delay the course of this disease with the ultimate goal of preventing neuron losses and preserving memory functioning. In this review we describe the renin-angiotensin II (AngII) system (RAS) with specific regard to its deleterious contributions to hypertension, facilitation of neuroinflammation and oxidative stress, reduced cerebral blood flow, tissue remodeling, and disruption of memory consolidation and retrieval. There is evidence that components of the RAS, AngIV and Ang(1-7), are positioned to counter such damaging influences and these systems are detailed with the goal of drawing attention to their importance as drug development targets. Ang(1-7) binds at the Mas receptor, while AngIV binds at the AT4 receptor subtype, and these receptor numbers are significantly decreased in AD patients, accompanied by declines in brain aminopeptidases A and N, enzymes essential for the synthesis of AngIV. Potent analogs may be useful to counter these changes and facilitate neuronal functioning and reduce apoptosis in memory associated brain structures of AD patients.
Collapse
Affiliation(s)
- John W Wright
- Department of Psychology, Washington State University, Pullman, WA, USA.,Department of Integrative Physiology and Neuroscience, and Program in Biotechnology, Washington State University, Pullman, WA, USA.,M3 Biotechnology, Inc., Seattle, WA, USA
| | - Joseph W Harding
- Department of Psychology, Washington State University, Pullman, WA, USA.,Department of Integrative Physiology and Neuroscience, and Program in Biotechnology, Washington State University, Pullman, WA, USA.,M3 Biotechnology, Inc., Seattle, WA, USA
| |
Collapse
|
10
|
Evans CE, Miners JS, Piva G, Willis CL, Heard DM, Kidd EJ, Good MA, Kehoe PG. ACE2 activation protects against cognitive decline and reduces amyloid pathology in the Tg2576 mouse model of Alzheimer's disease. Acta Neuropathol 2020; 139:485-502. [PMID: 31982938 PMCID: PMC7035243 DOI: 10.1007/s00401-019-02098-6] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 10/16/2019] [Accepted: 11/08/2019] [Indexed: 02/07/2023]
Abstract
Mid-life hypertension and cerebrovascular dysfunction are associated with increased risk of later life dementia, including Alzheimer’s disease (AD). The classical renin–angiotensin system (cRAS), a physiological regulator of blood pressure, functions independently within the brain and is overactive in AD. cRAS-targeting anti-hypertensive drugs are associated with reduced incidence of AD, delayed onset of cognitive decline, and reduced levels of Aβ and tau in both animal models and human pathological studies. cRAS activity is moderated by a downstream regulatory RAS pathway (rRAS), which is underactive in AD and is strongly associated with pathological hallmarks in human AD, and cognitive decline in animal models of CNS disease. We now show that enhancement of brain ACE2 activity, a major effector of rRAS, by intraperitoneal administration of diminazene aceturate (DIZE), an established activator of ACE2, lowered hippocampal Aβ and restored cognition in mid-aged (13–14-month-old) symptomatic Tg2576 mice. We confirmed that the protective effects of DIZE were directly mediated through ACE2 and were associated with reduced hippocampal soluble Aβ42 and IL1-β levels. DIZE restored hippocampal MasR levels in conjunction with increased NMDA NR2B and downstream ERK signalling expression in hippocampal synaptosomes from Tg2576 mice. Chronic (10 weeks) administration of DIZE to pre-symptomatic 9–10-month-old Tg2576 mice, and acute (10 days) treatment in cognitively impaired 12–13-month-old mice, prevented the development of cognitive impairment. Together these data demonstrate that ACE2 enhancement protects against and reverses amyloid-related hippocampal pathology and cognitive impairment in a preclinical model of AD.
Collapse
|
11
|
Telmisartan/17β-estradiol mitigated cognitive deficit in an ovariectomized rat model of Alzheimer's disease: Modulation of ACE1/ACE2 and AT1/AT2 ratio. Life Sci 2020; 245:117388. [PMID: 32007576 DOI: 10.1016/j.lfs.2020.117388] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 01/22/2020] [Accepted: 01/29/2020] [Indexed: 12/29/2022]
Abstract
AIMS The higher incidence rate of Alzheimer's disease (AD) among women has led to explorations on the association between estrogen deficiency and AD. Also, usage of antihypertensive drugs has been suggested to reduce the incidence of AD in elderly hypertensive patients. Thus, this study aimed to investigate the effects of telmisartan and/or 17β-estradiol on a cognitively impaired ovariectomized rat model of AD. MAIN METHODS 75 female Wistar rats were randomly allocated into five groups. One group was sham operated and the other four groups were subjected to ovariectomy, received D-galactose and either untreated or treated with telmisartan and/or 17β-estradiol for 6 weeks. KEY FINDINGS Ovariectomized rats showed cognitive impairment in Morris water maze and novel object recognition tests, increasing inflammatory biomarkers (tumor necrosis factor-α, and interleukin-1β), increasing AD biomarkers (amyloid beta1-42, and acetylcholine esterase), and over activation of classical arm of renin angiotensin system (RAS) (ACE1/Ang2/AT1) in hippocampi. Also, hippocampi histopathological examination revealed amyloid beta deposition. Whereas, administration of telmisartan and/or 17β-estradiol improved animals' behavior, alleviated histopathological alterations and reduced the level of inflammatory and AD biomarkers, modulated RAS activity favoring the novel neuroprotective arm (ACE2/Ang(1-7)/MasR). SIGNIFICANCE Our findings suggest that combined administration of both drugs has synergetic neuroprotective effects; supporting their potential application in AD treatment.
Collapse
|
12
|
Souza LAC, Trebak F, Kumar V, Satou R, Kehoe PG, Yang W, Wharton W, Feng Earley Y. Elevated cerebrospinal fluid sodium in hypertensive human subjects with a family history of Alzheimer's disease. Physiol Genomics 2020; 52:133-142. [PMID: 31961762 DOI: 10.1152/physiolgenomics.00093.2019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
High salt (sodium) intake leads to the development of hypertension despite the fact that plasma sodium concentration ([Na+]) is usually normal in hypertensive human patients. Increased cerebrospinal fluid (CSF) sodium contributes to elevated sympathetic activity and high blood pressure (BP) in rodent models of hypertension. However, whether there is an increased accumulation of sodium in the CSF of humans with chronic hypertension is not well defined. Here, we investigated CSF [Na+] from hypertensive and normotensive human subjects with family histories of Alzheimer's disease in samples collected in a clinical trial, as spinal tap is not a routine clinical procedure for hypertensive patients. The [Na+] and osmolality in plasma and CSF were measured by flame photometry. Daytime ambulatory BP was monitored while individuals were awake. Participants were deidentified and data were analyzed in conjunction with a retrospective analysis of patient history and diagnosis. We found that CSF [Na+] was significantly higher in participants with high BP compared with normotensive participants; there was no difference in plasma [Na+], or plasma and CSF osmolality between groups. Subsequent multiple linear regression analyses controlling for age, sex, race, and body mass index revealed a significant positive correlation between CSF [Na+] and BP but showed no correlation between plasma [Na+] and BP. In sum, CSF [Na+] was higher in chronic hypertensive individuals and may play a key role in the pathogenesis of human hypertension. Collectively, our findings provide evidence for the clinical significance of CSF [Na+] in chronic hypertension in humans.
Collapse
Affiliation(s)
- Lucas A C Souza
- Departments of Pharmacology and Physiology & Cell Biology, University of Nevada, Reno, School of Medicine, Reno, Nevada.,Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada
| | - Fatima Trebak
- Departments of Pharmacology and Physiology & Cell Biology, University of Nevada, Reno, School of Medicine, Reno, Nevada.,Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada
| | - Veena Kumar
- Department of Neurology, Emory University School of Nursing, Atlanta, Georgia
| | - Ryousuke Satou
- Department of Physiology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Patrick G Kehoe
- Institute of Clinical Neurosciences, University of Bristol, Bristol, United Kingdom
| | - Wei Yang
- University of Nevada, Reno, School of Community Health Sciences, Reno, Nevada
| | - Whitney Wharton
- Department of Neurology, Emory University School of Nursing, Atlanta, Georgia
| | - Yumei Feng Earley
- Departments of Pharmacology and Physiology & Cell Biology, University of Nevada, Reno, School of Medicine, Reno, Nevada.,Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada
| |
Collapse
|
13
|
Yasar S, Varma VR, Harris GC, Carlson MC. Associations of Angiotensin Converting Enzyme-1 and Angiotensin II Blood Levels and Cognitive Function. J Alzheimers Dis 2019; 63:655-664. [PMID: 29660936 DOI: 10.3233/jad-170944] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Emerging evidence suggests a possible role of the renin angiotensin system in the pathophysiologic process of Alzheimer's disease, of which angiotensin converting enzyme-1 (ACE-1) and angiotensin II (ANGII) are important proteins. Few studies evaluated associations between blood ACE-1 and none between ANGII levels, and cognition. OBJECTIVE Our pilot study was aimed to examine associations between blood ACE-1 and ANG II levels and cognitive function in non-demented participants at baseline and over a 1-year period. METHODS 56 participants were included from the Brain Health Substudy of the Baltimore Experience Corps Study. Linear regression analysis, adjusting for confounders, was used to determine associations between baseline ACE-1 and ANGII, and baseline and 1-year follow-up measures of psychomotor and processing speed, executive function, verbal learning memory and working memory, and whether these associations were mediated by blood pressure. RESULTS Participants were predominantly female (75%), African-American (93%), with mean age of 67.8 years and education of 14.3 years. There were no associations between baseline ACE-1 or ANGII levels and cognitive function; however, there were significant association between baseline ACE-1 levels and 1-year follow-up Trail Making Test, Part A (β= 0.003, p = 0.04) and Digit Span (β= -0.001, p = 0.02). CONCLUSIONS In this cognitively intact sample, elevated ACE-1 levels were associated with worse processing speed and working memory after 1 year. Findings from this pilot study suggest that changes in the RAS are associated with alterations in cognitive function warranting further assessment of the role of RAS in neurodegenerative disorders.
Collapse
Affiliation(s)
- Sevil Yasar
- Department of Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Vijay R Varma
- National Institute on Aging, Intramural Research Program, Laboratory of Behavioral Neuroscience, Baltimore, MD, USA
| | | | - Michelle C Carlson
- Department of Mental Health, Johns Hopkins University, Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
14
|
Eggink E, Moll van Charante EP, van Gool WA, Richard E. A Population Perspective on Prevention of Dementia. J Clin Med 2019; 8:E834. [PMID: 31212802 PMCID: PMC6617301 DOI: 10.3390/jcm8060834] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 06/06/2019] [Accepted: 06/09/2019] [Indexed: 01/21/2023] Open
Abstract
The global number of people living with dementia is expected to increase to 130 million in 2050. Based on extensive evidence from observational studies, it is estimated that about 30% of dementia cases may be attributable to potentially modifiable risk factors. This suggests that interventions targeting these factors could perhaps delay or prevent the onset of dementia. Since the vast majority of people with dementia live in low- and middle-income countries, such interventions should preferably be easy and affordable to implement across a wide range of health care systems. However, to date, results from dementia prevention trials do not provide convincing evidence that treatment of these risk factors reduces the risk of dementia. The current paper aims to give an overview of available evidence for the potential for dementia prevention. In particular, we discuss methodological issues that might complicate the development of effective prevention interventions and explore the opportunities and challenges for future dementia prevention research. Currently, several ongoing and planned trials are testing the effect of multi-domain interventions on dementia risk in high-risk populations. It is desirable that future dementia strategies also target the wider population, through interventions on the individual, community, and population level, in order to constrain the growing prevalence of dementia worldwide.
Collapse
Affiliation(s)
- Esmé Eggink
- Department of General Practice, Amsterdam University Medical Center, University of Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands.
| | - Eric P Moll van Charante
- Department of General Practice, Amsterdam University Medical Center, University of Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands.
| | - Willem A van Gool
- Department of Neurology, Amsterdam University Medical Center, University of Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands.
| | - Edo Richard
- Department of Neurology, Amsterdam University Medical Center, University of Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands.
- Department of Neurology, Donders Institute for Brain, Behaviour and Cognition, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands.
| |
Collapse
|
15
|
Kehoe PG. The Coming of Age of the Angiotensin Hypothesis in Alzheimer's Disease: Progress Toward Disease Prevention and Treatment? J Alzheimers Dis 2019; 62:1443-1466. [PMID: 29562545 PMCID: PMC5870007 DOI: 10.3233/jad-171119] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
There is wide recognition of a complex association between midlife hypertension and cardiovascular disease and later development of Alzheimer’s disease (AD) and cognitive impairment. While significant progress has been made in reducing rates of mortality and morbidity due to cardiovascular disease over the last thirty years, progress towards effective treatments for AD has been slower. Despite the known association between hypertension and dementia, research into each disease has largely been undertaken in parallel and independently. Yet over the last decade and a half, the emergence of converging findings from pre-clinical and clinical research has shown how the renin angiotensin system (RAS), which is very important in blood pressure regulation and cardiovascular disease, warrants careful consideration in the pathogenesis of AD. Numerous components of the RAS have now been found to be altered in AD such that the multifunctional and potent vasoconstrictor angiotensin II, and similarly acting angiotensin III, are greatly altered at the expense of other RAS signaling peptides considered to contribute to neuronal and cognitive function. Collectively these changes may contribute to many of the neuropathological hallmarks of AD, as well as observed progressive deficiencies in cognitive function, while also linking elements of a number of the proposed hypotheses for the cause of AD. This review discusses the emergence of the RAS and its likely importance in AD, not only because of the multiple facets of its involvement, but also perhaps fortuitously because of the ready availability of numerous RAS-acting drugs, that could be repurposed as interventions in AD.
Collapse
Affiliation(s)
- Patrick Gavin Kehoe
- Dementia Research Group, Translational Health Sciences, Bristol Medical School, University of Bristol, Southmead Hospital, Bristol, UK
| |
Collapse
|
16
|
Wharton W, Goldstein FC, Tansey MG, Brown AL, Tharwani SD, Verble DD, Cintron A, Kehoe PG. Rationale and Design of the Mechanistic Potential of Antihypertensives in Preclinical Alzheimer's (HEART) Trial. J Alzheimers Dis 2019; 61:815-824. [PMID: 29254080 DOI: 10.3233/jad-161198] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Research indicates that certain antihypertensive medications alter Alzheimer's disease (AD) biomarkers in Caucasians. The renin angiotensin system (RAS) regulates blood pressure (BP) in the body and the brain and may directly influence AD biomarkers, including amyloid-β (Aβ) neuropathology, cerebral blood flow (CBF), and inflammatory markers. This hypothesis is supported by studies, including ours, showing that antihypertensives targeting the RAS reduce the risk and slow the progression of AD in Caucasians. While mounting evidence supports a protective role of RAS medications in Caucasians, this mechanism has not been explored in African Americans. To assess the mechanism by which RAS medications modify the brain RAS, cerebrospinal fluid (CSF) Aβ, CBF, and inflammatory markers in African Americans, we are conducting an eight month, Phase Ib randomized, placebo controlled trial, enrolling 60 middle-aged (45-70 years), non-demented individuals, at risk for AD by virtue of a parental history. Participants include normotensive and treated hypertensives that have never been exposed to a RAS medication. Participants are randomized (1 : 1:1) by gender and BP medication use (yes/no) to one of three groups: placebo, or 20 mg, or 40 mg telmisartan (Micardis), to determine the dose required to penetrate the CNS. Our overarching hypothesis is that, compared to placebo, both doses of telmisartan will penetrate the CNS and produce salutary, dose dependent effects on the brain RAS as well as CSF Aβ, CBF, and CSF inflammatory markers in African Americans, over eight months. This manuscript describes the trial rationale and design.
Collapse
Affiliation(s)
- Whitney Wharton
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Felicia C Goldstein
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Malú G Tansey
- Department of Physiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Alexandra L Brown
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Sonum D Tharwani
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Danielle D Verble
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Amarallys Cintron
- Department of Physiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Patrick G Kehoe
- Dementia Research Group, Faculty of Health Sciences, University of Bristol, Learning and Research, Southmead Hospital, Bristol, UK
| |
Collapse
|
17
|
Wharton W, Zhao L, Steenland K, Goldstein FC, Schneider JA, Barnes LL, Gearing M, Yasar S. Neurofibrillary Tangles and Conversion to Mild Cognitive Impairment with Certain Antihypertensives. J Alzheimers Dis 2019; 70:153-161. [PMID: 31177216 PMCID: PMC7297195 DOI: 10.3233/jad-190011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND Individuals taking renin angiotensin system (RAS) acting antihypertensives exhibit slower cognitive decline and are less likely to progress from mild cognitive impairment (MCI) to Alzheimer's disease (AD), but the mechanism remains unclear. OBJECTIVE We tested the hypothesis that individuals taking RAS acting antihypertensives exhibit less AD-related neuropathology and slower disease progression than individuals taking non-RAS acting antihypertensives. METHOD Participants included 83 individuals with MCI who were taking an antihypertensive at baseline, had at least two follow-up visits, and had postmortem neuropathological data. Participants were old (M = 83.1 years), 32% male, well educated (M = 15.7 years), and 9.2% Black. RESULTS RAS medication users (N = 38) were less likely to progress to AD than non-RAS users (N = 45). RAS users exhibited fewer neurofibrillary tangles than non-RAS users in the hippocampal CA1 region (p < 0.01), entorhinal cortex (p = 0.03), and the angular gyrus, inferior temporal, mid-frontal cortex, and superior frontal (p = 0.01). CONCLUSION Prevention or clearance of neurofibrillary tangles represents a mechanism by which RAS medications may slow disease progression.
Collapse
Affiliation(s)
- Whitney Wharton
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
- Emory University, School of Nursing, Atlanta, GA, USA
| | - Liping Zhao
- Emory University Department of Biostatistics and Bioinformatics, School of Public Health, Atlanta, GA, USA
| | - Kyle Steenland
- Rollins School of Public Health, Biostatistics and Bioinformatics, Atlanta, GA, USA
| | | | - Julie A. Schneider
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Lisa L. Barnes
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Marla Gearing
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Sevil Yasar
- Johns Hopkins University School of Medicine, Department of Medicine, Baltimore, MD, USA
| |
Collapse
|
18
|
Hu Z, Wang L, Ma S, Kirisci L, Feng Z, Xue Y, Klunk WE, Kamboh MI, Sweet RA, Becker J, Lv Q, Lopez OL, Xie XQ. Synergism of antihypertensives and cholinesterase inhibitors in Alzheimer's disease. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2018; 4:542-555. [PMID: 30386819 PMCID: PMC6205113 DOI: 10.1016/j.trci.2018.09.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION We investigated the effect of antihypertensive (aHTN) medications and cholinesterase inhibitors (ChEIs) on the cognitive decline in patients with Alzheimer's disease (AD) and analyzed synergism by chemogenomics systems pharmacology mapping. METHODS We compared the effect of aHTN drugs on Mini-Mental State Examination scores in 617 AD patients with hypertension, and studied the synergistic effects. RESULTS The combination of diuretics, calcium channel blockers, and renin-angiotensin-aldosterone system blockers showed slower cognitive decline compared with other aHTN groups (Δβ = +1.46, P < .0001). aHTN medications slow down cognitive decline in ChEI users (Δβ = +0.56, P = .006), but not in non-ChEI users (Δβ = -0.31, P = .53). DISCUSSION aHTN and ChEI drugs showed synergistic effects. A combination of diuretics, renin-angiotensin-aldosterone system blockers, and calcium channel blockers had the slowest cognitive decline. The chemogenomics systems pharmacology-identified molecular targets provide system pharmacology interpretation of the synergism of the drugs in clinics. The results suggest that improving vascular health is essential for AD treatment and provide a novel direction for AD drug development.
Collapse
Affiliation(s)
- Ziheng Hu
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy; NIDA National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Lirong Wang
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy; NIDA National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Shifan Ma
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy; NIDA National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Levent Kirisci
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy; NIDA National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Zhiwei Feng
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy; NIDA National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ying Xue
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy; NIDA National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Clinical Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - William E. Klunk
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - M. Ilyas Kamboh
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Robert A. Sweet
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - James Becker
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Qianzhou Lv
- Department of Clinical Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Oscar L. Lopez
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xiang-Qun Xie
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy; NIDA National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
19
|
Lin YT, Wu YC, Sun GC, Ho CY, Wong TY, Lin CH, Chen HH, Yeh TC, Li CJ, Tseng CJ, Cheng PW. Effect of Resveratrol on Reactive Oxygen Species-Induced Cognitive Impairment in Rats with Angiotensin II-Induced Early Alzheimer's Disease †. J Clin Med 2018; 7:329. [PMID: 30301188 PMCID: PMC6210584 DOI: 10.3390/jcm7100329] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 09/28/2018] [Accepted: 10/02/2018] [Indexed: 01/08/2023] Open
Abstract
Recent studies have indicated that several anti-hypertensive drugs may delay the development and progression of Alzheimer's disease (AD). However, the relationships among AD, hypertension, and oxidative stress remain to be elucidated. Here, we aimed to determine whether reactive oxygen species (ROS) reduction by resveratrol in the brain leads to cognitive impairment reduction in rats with angiotensin II (Ang-II)-induced early AD. Male Wistar Kyoto (WKY) rats with Ang-II-induced AD were treated with losartan or resveratrol for two weeks. Our results show decreased blood pressure, increased hippocampal brain-derived neurotrophic factor (BDNF) level, and decreased nucleus tractus solitarius (NTS) ROS production in the Ang-II groups with losartan (10 mg/kg), or resveratrol (10 mg/kg/day) treatment. Furthermore, losartan inhibition of hippocampal TauT231 phosphorylation activated AktS473 phosphorylation, and significantly abolished Ang-II-induced Aβ precursors, active caspase 3, and glycogen synthase kinase 3β (GSK-3β)Y216 expressions. Consistently, resveratrol showed similar effects compared to losartan. Both losartan and resveratrol restored hippocampal-dependent contextual memory by NADPH oxidase 2 (NOX2) deletion and superoxide dismutase 2 (SOD2) elevation. Our results suggest that both losartan and resveratrol exert neuroprotective effects against memory impairment and hippocampal damage by oxidative stress reduction in early stage AD rat model. These novel findings indicate that resveratrol may represent a pharmacological option similar to losartan for patients with hypertension at risk of AD during old age.
Collapse
Affiliation(s)
- Yu-Te Lin
- Section of Neurology, Kaohsiung Veterans General Hospital, Kaohsiung 81300, Taiwan.
- Center for Geriatrics and Gerontology, Kaohsiung Veterans General Hospital, Kaohsiung 81300, Taiwan.
| | - Yi-Chung Wu
- Section of Neurology, Zouying Branch of Kaohsiung Armed Forces General Hospital Kaohsiung, Kaohsiung 81300, Taiwan.
| | - Gwo-Ching Sun
- Department of Anesthesiology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 81700, Taiwan.
| | - Chiu-Yi Ho
- Department of Biomedical Science, National Sun Yat-Sen University, Kaohsiung 80400, Taiwan.
| | - Tzyy-Yue Wong
- Research Assistant Center, Show Chwan Memorial Hospital, Changhua 50000, Taiwan.
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 81300, Taiwan.
| | - Ching-Huang Lin
- Section of Neurology, Kaohsiung Veterans General Hospital, Kaohsiung 81300, Taiwan.
| | - Hsin-Hung Chen
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 81300, Taiwan.
| | - Tung-Chen Yeh
- Department of Internal Medicine, Division of Cardiology, Kaohsiung Veterans General Hospital, Kaohsiung 81300, Taiwan.
| | - Chia-Jung Li
- Research Assistant Center, Show Chwan Memorial Hospital, Changhua 50000, Taiwan.
| | - Ching-Jiunn Tseng
- Department of Biomedical Science, National Sun Yat-Sen University, Kaohsiung 80400, Taiwan.
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 81300, Taiwan.
- Department of Pharmacology, Medical Research, China Medical University Hospital, China Medical University, Taichung 40400, Taiwan.
| | - Pei-Wen Cheng
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 81300, Taiwan.
- Yuh-Ing Junior College of Health Care & Management, Kaohsiung 82100, Taiwan.
- Shu-Zen Junior College of Medicine and Management, Kaohsiung 80700, Taiwan.
| |
Collapse
|
20
|
Kamel AS, Abdelkader NF, Abd El-Rahman SS, Emara M, Zaki HF, Khattab MM. Stimulation of ACE2/ANG(1–7)/Mas Axis by Diminazene Ameliorates Alzheimer’s Disease in the D-Galactose-Ovariectomized Rat Model: Role of PI3K/Akt Pathway. Mol Neurobiol 2018. [DOI: 10.1007/s12035-018-0966-3] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
21
|
Rius-Pérez S, Tormos A, Pérez S, Taléns-Visconti R. Patología vascular: ¿causa o efecto en la enfermedad de Alzheimer? Neurologia 2018; 33:112-120. [DOI: 10.1016/j.nrl.2015.07.010] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 07/28/2015] [Indexed: 10/23/2022] Open
|
22
|
Rius-Pérez S, Tormos A, Pérez S, Taléns-Visconti R. Vascular pathology: Cause or effect in Alzheimer disease? NEUROLOGÍA (ENGLISH EDITION) 2018. [DOI: 10.1016/j.nrleng.2015.07.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
23
|
Kehoe PG, Hibbs E, Palmer LE, Miners JS. Angiotensin-III is Increased in Alzheimer's Disease in Association with Amyloid-β and Tau Pathology. J Alzheimers Dis 2018; 58:203-214. [PMID: 28387670 DOI: 10.3233/jad-161265] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Hyperactivity of the renin-angiotensin system (RAS) is associated with the pathogenesis of Alzheimer's disease (AD) believed to be mediated by angiotensin-II (Ang-II) activation of the angiotensin type 1 receptor (AT1R). We previously showed that angiotensin-converting enzyme-1 (ACE-1) activity, the rate-limiting enzyme in the production of Ang-II, is increased in human postmortem brain tissue in AD. Angiotensin-III (Ang-III) activates the AT1R and angiotensin type-2 receptor (AT2R), but its potential role in the pathophysiology of AD remains unexplored. We measured Ang-II and Ang-III levels by ELISA, and the levels and activities of aminopeptidase-A (AP-A) and aminopeptidase-N (AP-N) (responsible for the production and metabolism of Ang-III, respectively) in human postmortem brain tissue in the mid-frontal cortex (Brodmann area 9) in a cohort of AD (n = 90) and age-matched non-demented controls (n = 59), for which we had previous measurements of ACE-1 activity, Aβ level, and tau pathology (also in the mid-frontal cortex). We found that both Ang-II and Ang-III levels were significantly higher in AD compared to age-matched controls and that Ang-III, rather than Ang-II, was strongly associated with Aβ load and tau load. Levels of AP-A were significantly reduced in AD but AP-A enzyme activity was unchanged whereas AP-N activity was reduced in AD but AP-N protein level was unchanged. Together, these data indicate that the APA/Ang-III/APN/Ang-IV/AT4R pathway is dysregulated and that elevated Ang-III could contribute to the pathogenesis of AD.
Collapse
|
24
|
Taheri S, Yu J, Zhu H, Kindy MS. High-Sodium Diet Has Opposing Effects on Mean Arterial Blood Pressure and Cerebral Perfusion in a Transgenic Mouse Model of Alzheimer's Disease. J Alzheimers Dis 2018; 54:1061-1072. [PMID: 27567835 DOI: 10.3233/jad-160331] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Cerebral ionic homeostasis impairment, especially Ca2+, has been observed in Alzheimer's disease (AD) and also with hypertension. Hypertension and AD both have been implicated in impaired cerebral autoregulation. However, the relationship between the ionic homeostasis impairment in AD and hypertension and cerebral blood flow (CBF) autoregulation is not clear. OBJECTIVE To test the hypothesis that a high-salt diet regimen influences the accumulation of amyloid-β (Aβand CBF) and CBF, exacerbates cognitive decline, and increases the propensity to AD. METHODS Double transgenic mice harboring the amyloid-β protein precursor (APPswe), and presenilin-1 (PSEN1) along with control littermates, 2 months of age at initiation of special diet, were divided into 4 groups: Group A, APP/PS1 and Group B, controls fed a high-sodium (4.00%) chow diet for 3 months; Group C, APP/PS1 and Group D, controls fed a low-sodium (0.08%) regular chow diet for 3 months. Mean arterial blood pressure (MAP) and CBF were measured noninvasively using the tail MAP measurement device and magnetic resonance imaging, respectively. Aβ plaques numbers in the cortex and hippocampus of APP/PS1 were quantified. RESULTS In contrary to controls, APP/PS1 mice fed a high-salt diet did not show markedly elevated mean systolic and diastolic blood pressure (134±4.8 compared with 162±2.8 mmHg, and 114±5.0 compared with 137±20 mmHg, p< 0.0001). However, a high-salt diet increased CBF in both APP/PS1 and controls and did not alter the cerebral tissue integrity. Aβ plaques were significantly reduced in the cortex and hippocampus of mice fed a high-salt diet. CONCLUSION These data suggest that a high-salt diet differently affects MAP and CBF in APP/PS1 mice and controls.
Collapse
Affiliation(s)
- Saeid Taheri
- Department of Pharmaceutical Sciences, University of South Florida, Tampa, FL, USA
| | - Jin Yu
- Department of Pharmaceutical Sciences, University of South Florida, Tampa, FL, USA
| | - Hong Zhu
- Department of Pharmaceutical Sciences, University of South Florida, Tampa, FL, USA
| | - Mark S Kindy
- Department of Pharmaceutical Sciences, University of South Florida, Tampa, FL, USA.,James A. Haley VA Medical Center, Tampa, FL, USA
| |
Collapse
|
25
|
Relationship Between Antihypertensive Medications and Cognitive Impairment: Part II. Review of Physiology and Animal Studies. Curr Hypertens Rep 2017; 18:66. [PMID: 27492369 PMCID: PMC4988998 DOI: 10.1007/s11906-016-0673-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
PURPOSE OF REVIEW There is an established association between hypertension and increased risk of poor cognitive performance and dementia including Alzheimer's disease; however, associations between antihypertensive medications (AHM) and dementia risk are less clear. An increased interest in AHM has resulted in expanding publications; however, none of the recent reviews provide comprehensive review. Our extensive review includes 24 mechanistic animal and human studies published over the last 5 years assessing relationship between AHM and cognitive function. RECENT FINDINGS All classes of AHM showed similar result patterns in animal studies. The mechanism by which AHM exert their effect was extensively studied by evaluating well-established pathways of AD disease process, including amyloid beta (Aβ), vascular, oxidative stress and inflammation pathways, but only few studies evaluated the blood pressure lowering effect on the AD disease process. Methodological limitations of the studies prevent comprehensive conclusions prior to further work evaluating AHM in animals and larger human observational studies, and selecting those with promising results for future RCTs.
Collapse
|
26
|
Mamelak M. Energy and the Alzheimer brain. Neurosci Biobehav Rev 2017; 75:297-313. [PMID: 28193453 DOI: 10.1016/j.neubiorev.2017.02.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 01/30/2017] [Accepted: 02/01/2017] [Indexed: 01/01/2023]
Abstract
The high energy demands of the poorly myelinated long axon hippocampal and cortical neurons render these neurons selectively vulnerable to degeneration in Alzheimer's disease. However, pathology engages all of the major elements of the neurovascular unit of the mature Alzheimer brain, the neurons, glia and blood vessels. Neurons present with retrograde degeneration of the axodendritic tree, capillaries with string vessels and markedly reduced densities and glia with signs of inflammatory activation. The neurons, capillaries and astrocytes of the mature Alzheimer brain harbor structurally defective mitochondria. Clinically, reduced glucose utilization, decades before cognitive deterioration, betrays ongoing energy insufficiency. β-hydroxybutyrate and γ-hydroxybutyrate can both provide energy to the brain when glucose utilization is blocked. Early work in mouse models of Alzheimer's disease demonstrate their ability to reverse the pathological changes in the Alzheimer brain and initial clinical trials reveal their ability to improve cognition and every day function. Supplying the brain with energy holds great promise for delaying the onset of Alzheimer's disease and slowing its progress.
Collapse
|
27
|
Arjmand Abbassi Y, Mohammadi MT, Sarami Foroshani M, Raouf Sarshoori J. Captopril and Valsartan May Improve Cognitive Function Through Potentiation of the Brain Antioxidant Defense System and Attenuation of Oxidative/Nitrosative Damage in STZ-Induced Dementia in Rat. Adv Pharm Bull 2016; 6:541-549. [PMID: 28101460 DOI: 10.15171/apb.2016.067] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 09/05/2016] [Accepted: 09/19/2016] [Indexed: 12/16/2022] Open
Abstract
Purpose: Previous findings have shown the crucial roles of brain renin-angiotensin system (RAS) in pathogenesis of Alzheimer's disease (AD). Since RAS inhibitors may have beneficial effects on dementia and cognitive function in elderly people, the aim of present study was to examine the neuroprotective actions of captopril and valsartan on memory function and neuronal damage in experimental model of AD. Methods: Adult forty male Wistar rats (220-280g) were randomly divided into 5 groups; Control, Vehicle, Alzheimer and treatment groups. AD was induced by the injections of streptozotocin (3mg/kg, bilateral intracerebroventricular) at days 1&3. Treated rats received orally captopril (50mg/kg/day) and valsartan (30mg/kg/day). Memory function and histological assessments were done at termination of experiment. Finally, superoxide dismutase (SOD) and catalase (CAT) activities as well as malondialdehyde (MDA) and NOx contents were determined. Results: There was a significant increase in the mean value of latency in Alzheimer group (66%). Captopril and valsartan considerably decreased this value in both treatment groups (45% and 72%, respectively). In Alzheimer group the activities of brain's SOD and CAT reduced (40% and 47%, respectively) in accompany with an increase in MDA and NOx contents (49% and 50%, respectively). Captopril and valsartan significantly increased the activities of brain's SOD and CAT concomitant reduction in MDA and NOx contents. Also, histopathological damages noticeably decreased in both treatment groups. Conclusion: Our findings indicate that RAS inhibition by using captopril and valsartan potentiates the antioxidant defense system of brain and reduces oxidative/nitrosative stress in accompany with neuronal damage during AD.
Collapse
Affiliation(s)
| | - Mohammad Taghi Mohammadi
- Department of Physiology and Biophysics, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mahsa Sarami Foroshani
- Department of Nanotechnology, School of new sciences and technology, Islamic Aazad University of Pharmaceutical Scinces Branch, Tehran, Iran
| | - Javad Raouf Sarshoori
- Department of Anatomy, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
28
|
Wang J, Tan L, Yu JT. Prevention Trials in Alzheimer's Disease: Current Status and Future Perspectives. J Alzheimers Dis 2016; 50:927-45. [PMID: 26836177 DOI: 10.3233/jad-150826] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia in the elderly. Over the past 20 years, both pharmacological and lifestyle interventions have been studied for AD prevention, but the overall results have been disappointing. The majority of disappointing results have raised questions and great challenges for the future of AD prevention trials. Ongoing advances in the knowledge of pathogenesis, in the identification of novel targets, in improved outcome measures, and in identification and validation of biomarkers may lead to effective strategies for AD prevention. In this paper, we review the selection of participants and interventions, trial design, outcome assessments, and promising biomarkers in prevention trials, and summarize the lessons learned from completed trials and perspectives from ongoing trials in AD prevention. Selection of optimal participants and interventions, coupled with more refined outcomes and more efficient trial design, may have the capacity to deliver a new era of preventive discovery in this challenging area.
Collapse
Affiliation(s)
- Jun Wang
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, China.,Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Yuzhong District, Chongqing, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, China
| | - Jin-Tai Yu
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA
| |
Collapse
|
29
|
Kehoe PG, Wong S, Al Mulhim N, Palmer LE, Miners JS. Angiotensin-converting enzyme 2 is reduced in Alzheimer's disease in association with increasing amyloid-β and tau pathology. ALZHEIMERS RESEARCH & THERAPY 2016; 8:50. [PMID: 27884212 PMCID: PMC5123239 DOI: 10.1186/s13195-016-0217-7] [Citation(s) in RCA: 142] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 10/20/2016] [Indexed: 12/14/2022]
Abstract
BACKGROUND Hyperactivity of the classical axis of the renin-angiotensin system (RAS), mediated by angiotensin II (Ang II) activation of the angiotensin II type 1 receptor (AT1R), is implicated in the pathogenesis of Alzheimer's disease (AD). Angiotensin-converting enzyme-2 (ACE-2) degrades Ang II to angiotensin 1-7 (Ang (1-7)) and counter-regulates the classical axis of RAS. We have investigated the expression and distribution of ACE-2 in post-mortem human brain tissue in relation to AD pathology and classical RAS axis activity. METHODS We measured ACE-2 activity by fluorogenic peptide substrate assay in mid-frontal cortex (Brodmann area 9) in a cohort of AD (n = 90) and age-matched non-demented controls (n = 59) for which we have previous data on ACE-1 activity, amyloid β (Aβ) level and tau pathology, as well as known ACE1 (rs1799752) indel polymorphism, apolipoprotein E (APOE) genotype, and cerebral amyloid angiopathy severity scores. RESULTS ACE-2 activity was significantly reduced in AD compared with age-matched controls (P < 0.0001) and correlated inversely with levels of Aβ (r = -0.267, P < 0.001) and phosphorylated tau (p-tau) pathology (r = -0.327, P < 0.01). ACE-2 was reduced in individuals possessing an APOE ε4 allele (P < 0.05) and was associated with ACE1 indel polymorphism (P < 0.05), with lower ACE-2 activity in individuals homozygous for the ACE1 insertion AD risk allele. ACE-2 activity correlated inversely with ACE-1 activity (r = -0.453, P < 0.0001), and the ratio of ACE-1 to ACE-2 was significantly elevated in AD (P < 0.0001). Finally, we show that the ratio of Ang II to Ang (1-7) (a proxy measure of ACE-2 activity indicating conversion of Ang II to Ang (1-7)) is reduced in AD. CONCLUSIONS Together, our findings indicate that ACE-2 activity is reduced in AD and is an important regulator of the central classical ACE-1/Ang II/AT1R axis of RAS, and also that dysregulation of this pathway likely plays a significant role in the pathogenesis of AD.
Collapse
Affiliation(s)
- Patrick Gavin Kehoe
- Dementia Research Group, University of Bristol, Level 1, Learning and Research, Southmead Hospital, Bristol, BS10 5NB, UK.
| | - Steffenny Wong
- Dementia Research Group, University of Bristol, Level 1, Learning and Research, Southmead Hospital, Bristol, BS10 5NB, UK
| | - Noura Al Mulhim
- Dementia Research Group, University of Bristol, Level 1, Learning and Research, Southmead Hospital, Bristol, BS10 5NB, UK
| | - Laura Elyse Palmer
- Dementia Research Group, University of Bristol, Level 1, Learning and Research, Southmead Hospital, Bristol, BS10 5NB, UK
| | - J Scott Miners
- Dementia Research Group, University of Bristol, Level 1, Learning and Research, Southmead Hospital, Bristol, BS10 5NB, UK.
| |
Collapse
|
30
|
Nation DA, Ho J, Yew B. Older Adults Taking AT1-Receptor Blockers Exhibit Reduced Cerebral Amyloid Retention. J Alzheimers Dis 2016; 50:779-89. [PMID: 26757036 DOI: 10.3233/jad-150487] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
BACKGROUND Evidence suggests that angiotensin II AT1-receptor blockers (ARBs) may be protective against dementia, and studies in transgenic animals indicate that this may be due to improved amyloid-β (Aβ) clearance. OBJECTIVE We investigated whether taking ARBs was associated with an attenuation of age-related increases in cerebral Aβ retention, and reduced progression to dementia. METHODS Eight hundred seventy-one stroke-free and dementia-free older adults from the Alzheimer's Disease Neuroimaging Initiative (ADNI) study underwent baseline lumbar puncture, and a subgroup (n = 124) underwent 12 and 24 month follow-up lumbar puncture. Participants were followed at variable intervals for clinical progression to dementia. Linear mixed models and ANCOVA compared ARBs users with those taking other antihypertensives (O-antiHTN) or no antihypertensives (No-antiHTN) on cerebrospinal fluid (CSF) Aβ and phosphorylated tau (P-tau) levels. Cox regression and chi-square analyses compared groups on progression to dementia. RESULTS ARBs users exhibited greater vascular risk and lower educational attainment than the No-antiHTN group. Longitudinal analyses indicated higher CSF Aβ and lower P-tau in ARBs users versus other groups. Cross-sectional analyses revealed age-related decreases in CSF Aβ in other groups but not ARBs users. ARBs users were less likely to progress to dementia and showed reduced rate of progression relative to the No-antiHTN group. DISCUSSION Patients taking ARBs showed an attenuation of age-related decreases in CSF Aβ, a finding that is consistent with studies done in transgenic animals. These findings may partly explain why ARBs users show reduced progression to dementia despite their lower educational attainment and greater vascular risk burden.
Collapse
|
31
|
|
32
|
Love S, Miners JS. Cerebrovascular disease in ageing and Alzheimer's disease. Acta Neuropathol 2016; 131:645-58. [PMID: 26711459 PMCID: PMC4835514 DOI: 10.1007/s00401-015-1522-0] [Citation(s) in RCA: 207] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Revised: 12/03/2015] [Accepted: 12/07/2015] [Indexed: 12/14/2022]
Abstract
Cerebrovascular disease (CVD) and Alzheimer’s disease (AD) have more in common than their association with ageing. They share risk factors and overlap neuropathologically. Most patients with AD have Aβ amyloid angiopathy and degenerative changes affecting capillaries, and many have ischaemic parenchymal abnormalities. Structural vascular disease contributes to the ischaemic abnormalities in some patients with AD. However, the stereotyped progression of hypoperfusion in this disease, affecting first the precuneus and cingulate gyrus, then the frontal and temporal cortex and lastly the occipital cortex, suggests that other factors are more important, particularly in early disease. Whilst demand for oxygen and glucose falls in late disease, functional MRI, near infrared spectroscopy to measure the saturation of haemoglobin by oxygen, and biochemical analysis of myelin proteins with differential susceptibility to reduced oxygenation have all shown that the reduction in blood flow in AD is primarily a problem of inadequate blood supply, not reduced metabolic demand. Increasing evidence points to non-structural vascular dysfunction rather than structural abnormalities of vessel walls as the main cause of cerebral hypoperfusion in AD. Several mediators are probably responsible. One that is emerging as a major contributor is the vasoconstrictor endothelin-1 (EDN1). Whilst there is clearly an additive component to the clinical and pathological effects of hypoperfusion and AD, experimental and clinical observations suggest that the disease processes also interact mechanistically at a cellular level in a manner that exacerbates both. The elucidation of some of the mechanisms responsible for hypoperfusion in AD and for the interactions between CVD and AD has led to the identification of several novel therapeutic approaches that have the potential to ameliorate ischaemic damage and slow the progression of neurodegenerative disease.
Collapse
Affiliation(s)
- Seth Love
- Institute of Clinical Neurosciences, School of Clinical Sciences, Learning and Research Level 2, Southmead Hospital, University of Bristol, Bristol, BS10 5NB, UK.
| | - J Scott Miners
- Institute of Clinical Neurosciences, School of Clinical Sciences, Learning and Research Level 2, Southmead Hospital, University of Bristol, Bristol, BS10 5NB, UK
| |
Collapse
|
33
|
Hamel E, Royea J, Ongali B, Tong XK. Neurovascular and Cognitive failure in Alzheimer's Disease: Benefits of Cardiovascular Therapy. Cell Mol Neurobiol 2016; 36:219-32. [PMID: 26993506 PMCID: PMC11482419 DOI: 10.1007/s10571-015-0285-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 10/06/2015] [Indexed: 12/19/2022]
Abstract
Alzheimer's disease (AD) is a multifactorial and multifaceted disease for which we currently have very little to offer since there is no curative therapy, with only limited disease-modifying drugs. Recent studies in AD mouse models that recapitulate the amyloid-β (Aβ) pathology converge to demonstrate that it is possible to salvage cerebrovascular function with a variety of drugs and, particularly, therapies used to treat cardiovascular diseases such as hypercholesterolemia and hypertension. These drugs can reestablish dilatory function mediated by various endothelial and smooth muscle ion channels as well as nitric oxide availability, benefits that result in normalized brain perfusion. These cerebrovascular benefits would favor brain perfusion, which may help maintain neuronal function and, possibly, delay cognitive failure. However, restoring cerebrovascular function in AD mouse models was not necessarily accompanied by rescue of cognitive deficits related to spatial learning and memory. The results with cardiovascular therapies rather suggest that drugs originally designed to treat cardiovascular diseases that concurrently restore cerebrovascular and cognitive function do so through their pleiotropic effects. Specifically, recent findings suggest that these drugs act directly on brain cells and neuronal pathways involved in memory formation, hence, working simultaneously albeit independently on neuronal and vascular targets. These findings may help select medications for patients with cardiovascular diseases at risk of developing AD with increasing age. Further, they may identify molecular targets for recovering memory pathways that bear potential for new therapeutic avenues.
Collapse
Affiliation(s)
- Edith Hamel
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, 3801 University Street, Suite 748, Montréal, QC, H3A 2B4, Canada.
| | - Jessika Royea
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, 3801 University Street, Suite 748, Montréal, QC, H3A 2B4, Canada
| | - Brice Ongali
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, 3801 University Street, Suite 748, Montréal, QC, H3A 2B4, Canada
| | - Xin-Kang Tong
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, 3801 University Street, Suite 748, Montréal, QC, H3A 2B4, Canada
| |
Collapse
|
34
|
Ashby EL, Miners JS, Kehoe PG, Love S. Effects of Hypertension and Anti-Hypertensive Treatment on Amyloid-β (Aβ) Plaque Load and Aβ-Synthesizing and Aβ-Degrading Enzymes in Frontal Cortex. J Alzheimers Dis 2016; 50:1191-203. [DOI: 10.3233/jad-150831] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
35
|
Elkahloun AG, Hafko R, Saavedra JM. An integrative genome-wide transcriptome reveals that candesartan is neuroprotective and a candidate therapeutic for Alzheimer's disease. ALZHEIMERS RESEARCH & THERAPY 2016; 8:5. [PMID: 26822027 PMCID: PMC4731966 DOI: 10.1186/s13195-015-0167-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2015] [Accepted: 12/08/2015] [Indexed: 12/21/2022]
Abstract
BACKGROUND Alzheimer's disease is the most frequent age-related dementia, and is currently without treatment. To identify possible targets for early therapeutic intervention we focused on glutamate excitotoxicity, a major early pathogenic factor, and the effects of candesartan, an angiotensin receptor blocker of neuroprotective efficacy in cell cultures and rodent models of Alzheimer's disease. The overall goal of the study was to determine whether gene analysis of drug effects in a primary neuronal culture correlate with alterations in gene expression in Alzheimer's disease, thus providing further preclinical evidence of beneficial therapeutic effects. METHODS Primary neuronal cultures were treated with candesartan at neuroprotective concentrations followed by excitotoxic glutamate amounts. We performed genome-wide expression profile analysis and data evaluation by ingenuity pathway analysis and gene set enrichment analysis, compared with alterations in gene expression from two independent published datasets identified by microarray analysis of postmortem hippocampus from Alzheimer's disease patients. Preferential expression in cerebrovascular endothelial cells or neurons was analyzed by comparison to published gene expression in these cells isolated from human cortex by laser capture microdissection. RESULTS Candesartan prevented glutamate upregulation or downregulation of several hundred genes in our cultures. Ingenuity pathway analysis and gene set enrichment analysis revealed that inflammation, cardiovascular disease and diabetes signal transduction pathways and amyloid β metabolism were major components of the neuronal response to glutamate excitotoxicity. Further analysis showed associations of glutamate-induced changes in the expression of several hundred genes, normalized by candesartan, with similar alterations observed in hippocampus from Alzheimer's disease patients. Gene analysis of neurons and cerebrovascular endothelial cells obtained by laser capture microdissection revealed that genes up- and downregulated by glutamate were preferentially expressed in endothelial cells and neurons, respectively. CONCLUSIONS Our data may be interpreted as evidence of direct candesartan neuroprotection beyond its effects on blood pressure, revealing common and novel disease mechanisms that may underlie the in vitro gene alterations reported here and glutamate-induced cell injury in Alzheimer's disease. Our observations provide novel evidence for candesartan neuroprotection through early molecular mechanisms of injury in Alzheimer's disease, supporting testing this compound in controlled clinical studies in the early stages of the illness.
Collapse
Affiliation(s)
- Abdel G Elkahloun
- Comparative genomics and Cancer Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Roman Hafko
- Section on Pharmacology, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Juan M Saavedra
- Section on Pharmacology, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA. .,Department of Pharmacology and Physiology, Georgetown University Medical Center, SE402 Med/Dent, 3900 Reservoir Road, Washington, DC, 20057, USA.
| |
Collapse
|
36
|
Andrieu S, Coley N, Lovestone S, Aisen PS, Vellas B. Prevention of sporadic Alzheimer's disease: lessons learned from clinical trials and future directions. Lancet Neurol 2015. [PMID: 26213339 DOI: 10.1016/s1474-4422(15)00153-2] [Citation(s) in RCA: 196] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Interventions that have even quite modest effects at the individual level could drastically reduce the future burden of dementia associated with Alzheimer's disease at the population level. In the past three decades, both pharmacological and lifestyle interventions have been studied for the prevention of cognitive decline or dementia in randomised controlled trials of individuals mostly aged older than 50-55 years with or without risk factors for Alzheimer's disease. Several trials testing the effects of physical activity, cognitive training, or antihypertensive interventions showed some evidence of efficacy on a primary cognitive endpoint. However, most of these trials had short follow-up periods, and further evidence is needed to confirm effectiveness and establish the optimum design or dose of interventions and ideal target populations. Important innovations in ongoing trials include the development of multidomain interventions, and the use of biomarker or genetic inclusion criteria. Challenges include the use of adaptive trial designs, the development of standardised, sensitive outcome measures, and the need for interventions that can be implemented in resource-poor settings.
Collapse
Affiliation(s)
- Sandrine Andrieu
- Inserm UMR1027, F-31073, Toulouse, France; University Toulouse III, Toulouse, France; Department of Epidemiology and Public Health, CHU Toulouse, Toulouse, France.
| | - Nicola Coley
- Inserm UMR1027, F-31073, Toulouse, France; University Toulouse III, Toulouse, France; Department of Epidemiology and Public Health, CHU Toulouse, Toulouse, France
| | | | - Paul S Aisen
- Alzheimer's Therapeutic Research Institute, University of Southern California, San Diego, CA, USA
| | - Bruno Vellas
- Inserm UMR1027, F-31073, Toulouse, France; University Toulouse III, Toulouse, France; Department of Geriatric Medicine, CHU Toulouse, Toulouse, France
| |
Collapse
|
37
|
Abstract
Alzheimer's disease (AD) is the most common form of dementia and is a major cause of disability and dependency amongst older people. AD drugs approved so far are symptomatic treatments and are not thought to affect the underlying disease process. Trials conducted with agents aiming to slow or stop disease progression in patients with AD have all failed, perhaps because they were tested too late in the disease process. Therefore, there has been a move towards prevention of AD. This paper presents an overview of trials testing pharmacological interventions for sporadic AD prevention. Those tested to date were initially developed for the treatment of AD or for the treatment of other conditions, rather than being specifically developed for AD prevention. Associated issues, such as evidence of 'proof-of-concept,' doses and safety, are discussed. A major shift has taken place in the methodology of AD prevention trials since the results of the first trials were published in the 1990s. New directions that are currently being considered in ongoing or future prevention trials are discussed, in terms of endpoints, target populations, and study design. The use of AD-specific drugs to prevent AD in high-risk individuals is currently limited by a lack of validated predictive and surrogate markers. Population approaches, such as lifestyle changes, are an alternative strategy that could be of public health interest, but may provide only limited benefits for individuals. The best chance of preventing AD may come from a combination of individual and population prevention approaches.
Collapse
|
38
|
Ye R, Hu Y, Yao A, Yang Y, Shi Y, Jiang Y, Zhang J. Impact of renin-angiotensin system-targeting antihypertensive drugs on treatment of Alzheimer's disease: a meta-analysis. Int J Clin Pract 2015; 69:674-81. [PMID: 25721930 DOI: 10.1111/ijcp.12626] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
OBJECTIVE The impacts of renin-angiotensin system (RAS)-targeting antihypertensive drugs on Alzheimer's disease (AD) remain controversial. We performed this meta-analysis to evaluate the precise value of RAS-targeting antihypertensive drugs in terms of attenuating incidence of AD and slowing down cognitive decline in patients with AD. METHODS A systematic literature search was conducted using PubMed, Embase, ScienceDirect and CBM (China Biology Medicine Disc) before September 2014. Studies analysing incidence of AD and cognitive changes in AD patients with RAS-targeting antihypertensive drugs were identified. The principal outcome measures were hazard ratios (HRs) for incidence of AD and standardised mean difference (SMD) for cognitive changes in AD patients. Pooled data were calculated using fixed or random effects models according to the heterogeneity. RESULTS In total, 12 studies involving 896,410 participants met our inclusion criteria. RAS-targeting antihypertensive drugs were significantly associated with a reduced incidence rate of AD (HR 0.81, 95% CI 0.72-0.92, p = 0.001). In subgroup analysis, both angiotensin renin blockers and angiotensin converting enzyme inhibitors were shown to effectively decrease the incidence rate of AD. In the analysis of cognitive changes, a slower rate of cognitive decline was observed in AD patients with RAS-targeting antihypertensive drug (SMD 0.30, 95% CI 0.09-0.50, p = 0.004), when randomised trials and observational trials were combined. However, analysis of randomised trials alone did not show the same result (SMD 0.20, 95% CI -0.10 to 0.50, p = 0.182). CONCLUSIONS Renin-angiotensin system-targeting antihypertensive drugs may be a potential treatment for reducing the incidence and progression of AD. Further studies on RAS-targeting antihypertensive drugs, especially large randomised clinical trials, should be conducted in the future.
Collapse
Affiliation(s)
- R Ye
- Department of Neurology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Y Hu
- Department of Neurology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - A Yao
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Y Yang
- Department of Neurology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Y Shi
- Department of Neurology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Y Jiang
- Department of Neurology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - J Zhang
- Department of Neurology, Zhongnan Hospital, Wuhan University, Wuhan, China
| |
Collapse
|
39
|
Wright JW, Kawas LH, Harding JW. The development of small molecule angiotensin IV analogs to treat Alzheimer's and Parkinson's diseases. Prog Neurobiol 2014; 125:26-46. [PMID: 25455861 DOI: 10.1016/j.pneurobio.2014.11.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 11/17/2014] [Accepted: 11/19/2014] [Indexed: 02/07/2023]
Abstract
Alzheimer's (AD) and Parkinson's (PD) diseases are neurodegenerative diseases presently without effective drug treatments. AD is characterized by general cognitive impairment, difficulties with memory consolidation and retrieval, and with advanced stages episodes of agitation and anger. AD is increasing in frequency as life expectancy increases. Present FDA approved medications do little to slow disease progression and none address the underlying progressive loss of synaptic connections and neurons. New drug design approaches are needed beyond cholinesterase inhibitors and N-methyl-d-aspartate receptor antagonists. Patients with PD experience the symptomatic triad of bradykinesis, tremor-at-rest, and rigidity with the possibility of additional non-motor symptoms including sleep disturbances, depression, dementia, and autonomic nervous system failure. This review summarizes available information regarding the role of the brain renin-angiotensin system (RAS) in learning and memory and motor functions, with particular emphasis on research results suggesting a link between angiotensin IV (AngIV) interacting with the AT4 receptor subtype. Currently there is controversy over the identity of this AT4 receptor protein. Albiston and colleagues have offered convincing evidence that it is the insulin-regulated aminopeptidase (IRAP). Recently members of our laboratory have presented evidence that the brain AngIV/AT4 receptor system coincides with the brain hepatocyte growth factor/c-Met receptor system. In an effort to resolve this issue we have synthesized a number of small molecule AngIV-based compounds that are metabolically stable, penetrate the blood-brain barrier, and facilitate compromised memory and motor systems. These research efforts are described along with details concerning a recently synthesized molecule, Dihexa that shows promise in overcoming memory and motor dysfunctions by augmenting synaptic connectivity via the formation of new functional synapses.
Collapse
Affiliation(s)
- John W Wright
- Departments of Psychology, Integrative Physiology and Neuroscience and Program in Biotechnology, Washington State University, Pullman, WA 99164-4820, USA; M3 Biotechnology, Inc., 4000 Mason Rd Suite 300, Box 352141, Seattle, WA 98195-2141, USA.
| | - Leen H Kawas
- Departments of Psychology, Integrative Physiology and Neuroscience and Program in Biotechnology, Washington State University, Pullman, WA 99164-4820, USA; M3 Biotechnology, Inc., 4000 Mason Rd Suite 300, Box 352141, Seattle, WA 98195-2141, USA
| | - Joseph W Harding
- Departments of Psychology, Integrative Physiology and Neuroscience and Program in Biotechnology, Washington State University, Pullman, WA 99164-4820, USA; M3 Biotechnology, Inc., 4000 Mason Rd Suite 300, Box 352141, Seattle, WA 98195-2141, USA
| |
Collapse
|
40
|
Cotter JD, Thornton SN, Lee JK, Laursen PB. Are we being drowned in hydration advice? Thirsty for more? EXTREME PHYSIOLOGY & MEDICINE 2014; 3:18. [PMID: 25356197 PMCID: PMC4212586 DOI: 10.1186/2046-7648-3-18] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 10/01/2014] [Indexed: 12/29/2022]
Abstract
Hydration pertains simplistically to body water volume. Functionally, however, hydration is one aspect of fluid regulation that is far more complex, as it involves the homeostatic regulation of total body fluid volume, composition and distribution. Deliberate or pathological alteration of these regulated factors can be disabling or fatal, whereas they are impacted by exercise and by all environmental stressors (e.g. heat, immersion, gravity) both acutely and chronically. For example, dehydration during exercising and environmental heat stress reduces water volume more than electrolyte content, causing hyperosmotic hypohydration. If exercise continues for many hours with access to food and water, composition returns to normal but extracellular volume increases well above baseline (if exercising upright and at low altitude). Repeating bouts of exercise or heat stress does likewise. Dehydration due to physical activity or environmental heat is a routine fluid-regulatory stress. How to gauge such dehydration and - more importantly-what to do about it, are contested heavily within sports medicine and nutrition. Drinking to limit changes in body mass is commonly advocated (to maintain ≤2% reduction), rather than relying on behavioural cues (mainly thirst) because the latter has been deemed too insensitive. This review, as part of the series on moving in extreme environments, critiques the validity, problems and merits of externally versus autonomously controlled fluid-regulatory behaviours, both acutely and chronically. Our contention is that externally advocated hydration policies (especially based on change in body mass with exercise in healthy individuals) have limited merit and are extrapolated and imposed too widely upon society, at the expense of autonomy. More research is warranted to examine whether ad libitum versus avid drinking is beneficial, detrimental or neither in: acute settings; adapting for obligatory dehydration (e.g. elite endurance competition in the heat), and; development of chronic diseases that are associated with an extreme lack of environmental stress.
Collapse
Affiliation(s)
- James David Cotter
- Exercise and Environmental Physiology, School of Physical Education, Sport and Exercise Sciences, Division of Sciences, University of Otago, PO Box 56, Dunedin 9054, New Zealand
| | - Simon N Thornton
- Faculté de Médecine, Université de Lorraine, U 1116 -INSERM-UL, 9, Avenue de la forêt de Haye, CS50-184 - 54505 VANDŒUVRE, Les Nancy cedex, France
| | - Jason Kw Lee
- Defence Medical and Environmental Research Institute, DSO National Laboratories, Singapore ; Yong Loo Lin School of Medicine, National University of Singapore, Singapore ; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Paul B Laursen
- High Performance Sport New Zealand, Auckland, New Zealand ; Sports Performance Research Institute New Zealand (SPRINZ), Auckland University of Technology, Auckland, New Zealand
| |
Collapse
|
41
|
Miners JS, Palmer JC, Tayler H, Palmer LE, Ashby E, Kehoe PG, Love S. Aβ degradation or cerebral perfusion? Divergent effects of multifunctional enzymes. Front Aging Neurosci 2014; 6:238. [PMID: 25309424 PMCID: PMC4160973 DOI: 10.3389/fnagi.2014.00238] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 08/20/2014] [Indexed: 12/17/2022] Open
Abstract
There is increasing evidence that deficient clearance of β-amyloid (Aβ) contributes to its accumulation in late-onset Alzheimer disease (AD). Several Aβ-degrading enzymes, including neprilysin (NEP), endothelin-converting enzyme (ECE), and angiotensin-converting enzyme (ACE) reduce Aβ levels and protect against cognitive impairment in mouse models of AD. In post-mortem human brain tissue we have found that the activity of these Aβ-degrading enzymes rise with age and increases still further in AD, perhaps as a physiological response that helps to minimize the build-up of Aβ. ECE-1/-2 and ACE are also rate-limiting enzymes in the production of endothelin-1 (ET-1) and angiotensin II (Ang II), two potent vasoconstrictors, increases in the levels of which are likely to contribute to reduced blood flow in AD. This review considers the possible interdependence between Aβ-degrading enzymes, ischemia and Aβ in AD: ischemia has been shown to increase Aβ production both in vitro and in vivo, whereas increased Aβ probably enhances ischemia by vasoconstriction, mediated at least in part by increased ECE and ACE activity. In contrast, NEP activity may help to maintain cerebral perfusion, by reducing the accumulation of Aβ in cerebral blood vessels and lessening its toxicity to vascular smooth muscle cells. In assessing the role of Aβ-degrading proteases in the pathogenesis of AD and, particularly, their potential as therapeutic agents, it is important to bear in mind the multifunctional nature of these enzymes and to consider their effects on other substrates and pathways.
Collapse
Affiliation(s)
- J Scott Miners
- Dementia Research Group, School of Clinical Sciences, Faculty of Medicine and Dentistry, University of Bristol Bristol, UK
| | - Jennifer C Palmer
- Dementia Research Group, School of Clinical Sciences, Faculty of Medicine and Dentistry, University of Bristol Bristol, UK
| | - Hannah Tayler
- Dementia Research Group, School of Clinical Sciences, Faculty of Medicine and Dentistry, University of Bristol Bristol, UK
| | - Laura E Palmer
- Dementia Research Group, School of Clinical Sciences, Faculty of Medicine and Dentistry, University of Bristol Bristol, UK
| | - Emma Ashby
- Dementia Research Group, School of Clinical Sciences, Faculty of Medicine and Dentistry, University of Bristol Bristol, UK
| | - Patrick G Kehoe
- Dementia Research Group, School of Clinical Sciences, Faculty of Medicine and Dentistry, University of Bristol Bristol, UK
| | - Seth Love
- Dementia Research Group, School of Clinical Sciences, Faculty of Medicine and Dentistry, University of Bristol Bristol, UK
| |
Collapse
|
42
|
Cermakova P, Fereshtehnejad SM, Johnell K, Winblad B, Eriksdotter M, Religa D. Cardiovascular medication burden in dementia disorders: a nationwide study of 19,743 dementia patients in the Swedish Dementia Registry. ALZHEIMERS RESEARCH & THERAPY 2014; 6:34. [PMID: 25024749 PMCID: PMC4095690 DOI: 10.1186/alzrt264] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 06/03/2014] [Indexed: 12/29/2022]
Abstract
Introduction Administration of several cardiovascular drugs has an effect on dementia. We aimed to investigate whether there are differences in the use of cardiovascular medication between different dementia disorders. Methods We obtained information about dementia patients from the Swedish Dementia Registry. Patients were diagnosed with one of these dementia disorders: Alzheimer’s disease (n = 8,139), mixed dementia (n = 5,203), vascular dementia (n = 4,982), Lewy body dementia (n = 605), frontotemporal dementia (n = 409) and Parkinson’s disease dementia (n = 405). Multivariate logistic regression analysis was performed to investigate the association between use of cardiovascular medication and dementia disorders, after adjustment for age, gender, living alone, cognitive status and total number of drugs (a proxy for overall co-morbidity). Results Seventy percent of all the dementia patients used cardiovascular medication. Use of cardiovascular drugs is common in patients with vascular and mixed dementia. Male gender, higher age, slightly better cognitive status and living with another person was associated with use of cardiovascular medication. Conclusions Cardiovascular medication is used extensively across dementia disorders and particularly in vascular and mixed dementia. Future research should investigate the tolerability and effectiveness of these drugs in the different dementia disorders.
Collapse
Affiliation(s)
- Pavla Cermakova
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division for Neurogeriatrics, Karolinska Institutet, 141 57 Huddinge, Sweden ; International Clinical Research Center and St.Anne's University Hospital, Pekařská 53, 656 91 Brno, Czech Republic
| | - Seyed-Mohammad Fereshtehnejad
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Clinical Geriatrics, Karolinska Institutet, 141 57 Huddinge, Sweden
| | - Kristina Johnell
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Aging Research Center, Karolinska Institutet and Stockholm University, Gävlegatan 16, 113 30 Stockholm, Sweden
| | - Bengt Winblad
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division for Neurogeriatrics, Karolinska Institutet, 141 57 Huddinge, Sweden ; Department of Geriatric Medicine, Karolinska University Hospital, 141 86 Huddinge, Sweden
| | - Maria Eriksdotter
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division of Clinical Geriatrics, Karolinska Institutet, 141 57 Huddinge, Sweden ; Department of Geriatric Medicine, Karolinska University Hospital, 141 86 Huddinge, Sweden
| | - Dorota Religa
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Division for Neurogeriatrics, Karolinska Institutet, 141 57 Huddinge, Sweden ; Department of Geriatric Medicine, Karolinska University Hospital, 141 86 Huddinge, Sweden
| |
Collapse
|
43
|
Cacabelos R, Cacabelos P, Torrellas C, Tellado I, Carril JC. Pharmacogenomics of Alzheimer's disease: novel therapeutic strategies for drug development. Methods Mol Biol 2014; 1175:323-556. [PMID: 25150875 DOI: 10.1007/978-1-4939-0956-8_13] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is a major problem of health and disability, with a relevant economic impact on our society. Despite important advances in pathogenesis, diagnosis, and treatment, its primary causes still remain elusive, accurate biomarkers are not well characterized, and the available pharmacological treatments are not cost-effective. As a complex disorder, AD is a polygenic and multifactorial clinical entity in which hundreds of defective genes distributed across the human genome may contribute to its pathogenesis. Diverse environmental factors, cerebrovascular dysfunction, and epigenetic phenomena, together with structural and functional genomic dysfunctions, lead to amyloid deposition, neurofibrillary tangle formation, and premature neuronal death, the major neuropathological hallmarks of AD. Future perspectives for the global management of AD predict that genomics and proteomics may help in the search for reliable biomarkers. In practical terms, the therapeutic response to conventional drugs (cholinesterase inhibitors, multifactorial strategies) is genotype-specific. Genomic factors potentially involved in AD pharmacogenomics include at least five categories of gene clusters: (1) genes associated with disease pathogenesis; (2) genes associated with the mechanism of action of drugs; (3) genes associated with drug metabolism (phase I and II reactions); (4) genes associated with drug transporters; and (5) pleiotropic genes involved in multifaceted cascades and metabolic reactions. The implementation of pharmacogenomic strategies will contribute to optimize drug development and therapeutics in AD and related disorders.
Collapse
Affiliation(s)
- Ramón Cacabelos
- Chair of Genomic Medicine, Camilo José Cela University, 28692, Villanueva de la Cañada, Madrid, Spain,
| | | | | | | | | |
Collapse
|
44
|
O’Caoimh R, Kehoe PG, Kennedy P, Molloy W. Use of angiotensin-converting enzyme inhibitors for Alzheimer’s disease: an update. Neurodegener Dis Manag 2013. [DOI: 10.2217/nmt.13.61] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Rónán O’Caoimh
- Centre for Gerontology & Rehabilitation, University College Cork, St Finbarrs Hospital, Cork, Ireland
| | - Patrick Gavin Kehoe
- Dementia Research Group, School of Clinical Sciences, University of Bristol, Frenchay Hospital, Bristol, BS16 1LE, UK
| | - Paul Kennedy
- Centre for Gerontology & Rehabilitation, University College Cork, St Finbarrs Hospital, Cork, Ireland
| | - William Molloy
- Centre for Gerontology & Rehabilitation, University College Cork, St Finbarrs Hospital, Cork, Ireland
| |
Collapse
|