1
|
Bobkova NV, Chuvakova LN, Kovalev VI, Zhdanova DY, Chaplygina AV, Rezvykh AP, Evgen'ev MB. A Mouse Model of Sporadic Alzheimer's Disease with Elements of Major Depression. Mol Neurobiol 2024:10.1007/s12035-024-04346-7. [PMID: 38980563 DOI: 10.1007/s12035-024-04346-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 07/02/2024] [Indexed: 07/10/2024]
Abstract
After olfactory bulbectomy, animals are often used as a model of major depression or sporadic Alzheimer's disease and, hence, the status of this model is still disputable. To elucidate the nature of alterations in the expression of the genome after the operation, we analyzed transcriptomes of the cortex, hippocampus, and cerebellum of the olfactory bulbectomized (OBX) mice. Analysis of the functional significance of genes in the brain of OBX mice indicates that the balance of the GABA/glutamatergic systems is disturbed with hyperactivation of the latter in the hippocampus, leading to the development of excitotoxicity and induction of apoptosis in the background of severe mitochondrial dysfunction and astrogliosis. On top of this, the synthesis of neurotrophic factors decreases leading to the disruption of the cytoskeleton of neurons, an increase in the level of intracellular calcium, and the activation of tau protein hyperphosphorylation. Moreover, the acetylcholinergic system is deficient in the background of the hyperactivation of acetylcholinesterase. Importantly, the activity of the dopaminergic, endorphin, and opiate systems in OBX mice decreases, leading to hormonal dysfunction. On the other hand, genes responsible for the regulation of circadian rhythms, cell migration, and innate immunity are activated in OBX animals. All this takes place in the background of a drastic downregulation of ribosomal protein genes in the brain. The obtained results indicate that OBX mice represent a model of Alzheimer's disease with elements of major depression.
Collapse
Affiliation(s)
- N V Bobkova
- Institute of Cell Biophysics of the Russian Academy of Sciences-Federal Research Center, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, 142290, Pushchino, Moscow Region, Russia
| | - L N Chuvakova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991, Moscow, Russia
| | - V I Kovalev
- Institute of Cell Biophysics of the Russian Academy of Sciences-Federal Research Center, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, 142290, Pushchino, Moscow Region, Russia
| | - D Y Zhdanova
- Institute of Cell Biophysics of the Russian Academy of Sciences-Federal Research Center, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, 142290, Pushchino, Moscow Region, Russia
| | - A V Chaplygina
- Institute of Cell Biophysics of the Russian Academy of Sciences-Federal Research Center, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, 142290, Pushchino, Moscow Region, Russia
| | - A P Rezvykh
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991, Moscow, Russia
| | - M B Evgen'ev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991, Moscow, Russia.
| |
Collapse
|
2
|
Barak R, Goshtasbi G, Fatehi R, Firouzabadi N. Signaling pathways and genetics of brain Renin angiotensin system in psychiatric disorders: State of the art. Pharmacol Biochem Behav 2024; 236:173706. [PMID: 38176544 DOI: 10.1016/j.pbb.2023.173706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 12/19/2023] [Accepted: 12/28/2023] [Indexed: 01/06/2024]
Abstract
Along the conventional pathways, Renin-angiotensin system (RAS) plays a key role in the physiology of the CNS and pathogenesis of psychiatric diseases. RAS is a complex regulatory pathway which is composed of several peptides and receptors and comprises two counter-regulatory axes. The classical (ACE1/AngII/AT1 receptor) axis and the contemporary (ACE2/Ang (1-7)/Mas receptor) axis. The genes coding for elements of both axes have been broadly studied. Numerous functional polymorphisms on components of RAS have been identified to serve as informative disease and treatment markers. This review summarizes the role of each peptide and receptor in the pathophysiology of psychiatric disorders (depression, bipolar disorders and schizophrenia), followed by a concise look at the role of genetic polymorphism of the RAS in the pathophysiology of these disorders.
Collapse
Affiliation(s)
- Roya Barak
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Ghazal Goshtasbi
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reihaneh Fatehi
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negar Firouzabadi
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
3
|
Ayyubova G, Gychka SG, Nikolaienko SI, Alghenaim FA, Teramoto T, Shults NV, Suzuki YJ. The Role of Furin in the Pathogenesis of COVID-19-Associated Neurological Disorders. Life (Basel) 2024; 14:279. [PMID: 38398788 PMCID: PMC10890058 DOI: 10.3390/life14020279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 02/15/2024] [Indexed: 02/25/2024] Open
Abstract
Neurological disorders have been reported in a large number of coronavirus disease 2019 (COVID-19) patients, suggesting that this disease may have long-term adverse neurological consequences. COVID-19 occurs from infection by a positive-sense single-stranded RNA virus called severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The membrane fusion protein of SARS-CoV-2, the spike protein, binds to its human host receptor, angiotensin-converting enzyme 2 (ACE2), to initiate membrane fusion between the virus and host cell. The spike protein of SARS-CoV-2 contains the furin protease recognition site and its cleavage enhances the infectivity of this virus. The binding of SARS-CoV-2 to the ACE2 receptor has been shown to downregulate ACE2, thereby increasing the levels of pathogenic angiotensin II (Ang II). The furin protease cleaves between the S1 subunit of the spike protein with the binding domain toward ACE2 and the S2 subunit with the transmembrane domain that anchors to the viral membrane, and this activity releases the S1 subunit into the blood circulation. The released S1 subunit of the spike protein also binds to and downregulates ACE2, in turn increasing the level of Ang II. Considering that a viral particle contains many spike protein molecules, furin-dependent cleavage would release many free S1 protein molecules, each of which can downregulate ACE2, while infection with a viral particle only affects one ACE2 molecule. Therefore, the furin-dependent release of S1 protein would dramatically amplify the ability to downregulate ACE2 and produce Ang II. We hypothesize that this amplification mechanism that the virus possesses, but not the infection per se, is the major driving force behind COVID-19-associated neurological disorders.
Collapse
Affiliation(s)
- Gunel Ayyubova
- Department of Cytology, Embryology and Histology, Azerbaijan Medical University, Baku AZ1022, Azerbaijan
| | - Sergiy G Gychka
- Department of Pathological Anatomy, Bogomolets National Medical University, 01601 Kyiv, Ukraine
| | - Sofia I Nikolaienko
- Department of Pathological Anatomy, Bogomolets National Medical University, 01601 Kyiv, Ukraine
| | - Fada A Alghenaim
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC 20007, USA
| | - Tadahisa Teramoto
- Department of Microbiology and Immunology, Georgetown University Medical Center, Washington, DC 20007, USA
| | - Nataliia V Shults
- Department of Biology, Georgetown University, Washington, DC 20007, USA
| | - Yuichiro J Suzuki
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC 20007, USA
| |
Collapse
|
4
|
Reveret L, Leclerc M, Emond V, Tremblay C, Loiselle A, Bourassa P, Bennett DA, Hébert SS, Calon F. Higher angiotensin-converting enzyme 2 (ACE2) levels in the brain of individuals with Alzheimer's disease. Acta Neuropathol Commun 2023; 11:159. [PMID: 37784209 PMCID: PMC10544218 DOI: 10.1186/s40478-023-01647-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 08/30/2023] [Indexed: 10/04/2023] Open
Abstract
Cognitive decline due to Alzheimer's disease (AD) is frequent in the geriatric population, which has been disproportionately affected by the COVID-19 pandemic. In this study, we investigated the levels of angiotensin-converting enzyme 2 (ACE2), a regulator of the renin-angiotensin system and the main entry receptor of SARS-CoV-2 in host cells, in postmortem parietal cortex samples from two independent AD cohorts, totalling 142 persons. Higher concentrations of ACE2 protein (p < 0.01) and mRNA (p < 0.01) were found in individuals with a neuropathological diagnosis of AD compared to age-matched healthy control subjects. Brain levels of soluble ACE2 were inversely associated with cognitive scores (p = 0.02) and markers of pericytes (PDGFRβ, p = 0.02 and ANPEP, p = 0.007), but positively correlated with concentrations of soluble amyloid-β peptides (Aβ) (p = 0.01) and insoluble phospho-tau (S396/404, p = 0.002). However, no significant differences in ACE2 were observed in the 3xTg-AD mouse model of tau and Aβ neuropathology. Results from immunofluorescence and Western blots showed that ACE2 protein is predominantly localized in microvessels in the mouse brain whereas it is more frequently found in neurons in the human brain. The present data suggest that higher levels of soluble ACE2 in the human brain may contribute to AD, but their role in CNS infection by SARS-CoV-2 remains unclear.
Collapse
Affiliation(s)
- Louise Reveret
- Faculty of Pharmacy, Laval University, Quebec, QC, Canada
- CHU de Quebec Research Center, 2705, Boulevard Laurier, Room T2-05, Québec, QC, G1V 4G2, Canada
| | - Manon Leclerc
- Faculty of Pharmacy, Laval University, Quebec, QC, Canada
- CHU de Quebec Research Center, 2705, Boulevard Laurier, Room T2-05, Québec, QC, G1V 4G2, Canada
| | - Vincent Emond
- CHU de Quebec Research Center, 2705, Boulevard Laurier, Room T2-05, Québec, QC, G1V 4G2, Canada
| | - Cyntia Tremblay
- CHU de Quebec Research Center, 2705, Boulevard Laurier, Room T2-05, Québec, QC, G1V 4G2, Canada
| | - Andréanne Loiselle
- CHU de Quebec Research Center, 2705, Boulevard Laurier, Room T2-05, Québec, QC, G1V 4G2, Canada
| | - Philippe Bourassa
- Faculty of Pharmacy, Laval University, Quebec, QC, Canada
- CHU de Quebec Research Center, 2705, Boulevard Laurier, Room T2-05, Québec, QC, G1V 4G2, Canada
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Sébastien S Hébert
- CHU de Quebec Research Center, 2705, Boulevard Laurier, Room T2-05, Québec, QC, G1V 4G2, Canada
- Faculty of Medicine, Laval University, Quebec, QC, Canada
| | - Frédéric Calon
- Faculty of Pharmacy, Laval University, Quebec, QC, Canada.
- CHU de Quebec Research Center, 2705, Boulevard Laurier, Room T2-05, Québec, QC, G1V 4G2, Canada.
| |
Collapse
|
5
|
Villapol S, Janatpour ZC, Affram KO, Symes AJ. The Renin Angiotensin System as a Therapeutic Target in Traumatic Brain Injury. Neurotherapeutics 2023; 20:1565-1591. [PMID: 37759139 PMCID: PMC10684482 DOI: 10.1007/s13311-023-01435-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2023] [Indexed: 09/29/2023] Open
Abstract
Traumatic brain injury (TBI) is a major public health problem, with limited pharmacological options available beyond symptomatic relief. The renin angiotensin system (RAS) is primarily known as a systemic endocrine regulatory system, with major roles controlling blood pressure and fluid homeostasis. Drugs that target the RAS are used to treat hypertension, heart failure and kidney disorders. They have now been used chronically by millions of people and have a favorable safety profile. In addition to the systemic RAS, it is now appreciated that many different organ systems, including the brain, have their own local RAS. The major ligand of the classic RAS, Angiotensin II (Ang II) acts predominantly through the Ang II Type 1 receptor (AT1R), leading to vasoconstriction, inflammation, and heightened oxidative stress. These processes can exacerbate brain injuries. Ang II receptor blockers (ARBs) are AT1R antagonists. They have been shown in several preclinical studies to enhance recovery from TBI in rodents through improvements in molecular, cellular and behavioral correlates of injury. ARBs are now under consideration for clinical trials in TBI. Several different RAS peptides that signal through receptors distinct from the AT1R, are also potential therapeutic targets for TBI. The counter regulatory RAS pathway has actions that oppose those stimulated by AT1R signaling. This alternative pathway has many beneficial effects on cells in the central nervous system, bringing about vasodilation, and having anti-inflammatory and anti-oxidative stress actions. Stimulation of this pathway also has potential therapeutic value for the treatment of TBI. This comprehensive review will provide an overview of the various components of the RAS, with a focus on their direct relevance to TBI pathology. It will explore different therapeutic agents that modulate this system and assess their potential efficacy in treating TBI patients.
Collapse
Affiliation(s)
- Sonia Villapol
- Department of Neurosurgery, Houston Methodist Hospital, Houston, TX, USA
| | - Zachary C Janatpour
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Kwame O Affram
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Aviva J Symes
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
| |
Collapse
|
6
|
Puranik N, Yadav D, Song M. Advancements in the Application of Nanomedicine in Alzheimer's Disease: A Therapeutic Perspective. Int J Mol Sci 2023; 24:14044. [PMID: 37762346 PMCID: PMC10530821 DOI: 10.3390/ijms241814044] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/11/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease that affects most people worldwide. AD is a complex central nervous system disorder. Several drugs have been designed to cure AD, but with low success rates. Because the blood-brain and blood-cerebrospinal fluid barriers are two barriers that protect the central nervous system, their presence has severely restricted the efficacy of many treatments that have been studied for AD diagnosis and/or therapy. The use of nanoparticles for the diagnosis and treatment of AD is the focus of an established and rapidly developing field of nanomedicine. Recent developments in nanomedicine have made it possible to effectively transport drugs to the brain. However, numerous obstacles remain to the successful use of nanomedicines in clinical settings for AD treatment. Furthermore, given the rapid advancement in nanomedicine therapeutics, better outcomes for patients with AD can be anticipated. This article provides an overview of recent developments in nanomedicine using different types of nanoparticles for the management and treatment of AD.
Collapse
Affiliation(s)
| | | | - Minseok Song
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea; (N.P.); (D.Y.)
| |
Collapse
|
7
|
Müller JDA, Giacobe LJ, Grassi V, Palmeira ALR. The use of angiotensin receptor blockers in dementia prevention. Dement Neuropsychol 2023; 17:e20233006. [PMID: 37681194 PMCID: PMC10481907 DOI: 10.1590/1980-5764-dn-2023-3006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Accepted: 03/14/2023] [Indexed: 09/09/2023] Open
Abstract
Alzheimer's disease (AD) and dementia are preventable and highly prevalent diseases, as is systemic arterial hypertension. Thus, it is speculated that angiotensin receptor blockers (ARBs) may be neuroprotective against AD. Objective The aim of this study was to evaluate if the use of ARBs confers a neuroprotective effect on AD, through a systematic review. Methods Studies published on Embase, LILACS, SciELO, and PubMed were evaluated. The selection of the studies included those that evaluated the use of antihypertensive drugs in individuals with a previous diagnosis of mild cognitive impairment. The data were extracted with the Cochrane Effective Practice and Organization of Care (EPOC) form. The risk of bias was evaluated by the EPOC "Risk of bias tool." Results A total of 12 articles were identified, and 3 articles were selected. Two of them analyzed the use of ARB/ACEI versus other antihypertensives and the development of dementia. Conclusion There is a tendency for ARBs to be superior to other antihypertensives in preventing dementia.
Collapse
Affiliation(s)
| | | | - Vanise Grassi
- Universidade do Vale do Taquari, Centro de Ciências Médicas, Lajeado RS, Brazil
| | | |
Collapse
|
8
|
Yoshida T, Mori T, Shimizu H, Tachibana A, Yoshino Y, Ochi S, Yamazaki K, Ozaki Y, Kawabe K, Horiuchi F, Komori K, Iga JI, Ueno SI. Analysis of factors related to cognitive impairment in a community-based, complete enumeration survey in Japan: the Nakayama study. Psychogeriatrics 2023; 23:876-884. [PMID: 37483119 DOI: 10.1111/psyg.13012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 07/05/2023] [Accepted: 07/07/2023] [Indexed: 07/25/2023]
Abstract
BACKGROUND The number of patients with cognitive disorders is rapidly increasing in the world, becoming not only a medical problem, but also a social problem. There have been many reports that various factors are associated with cognitive dysfunction, but the factors have not yet been fully identified. This was a community-based complete enumeration study which aimed to identify risk and protective factors for dementia. METHODS The first phase included all residents aged 65 years or older in a town in Japan. They completed many examinations, such as living conditions questionnaires, physical examination, Mini-Mental State Examination, and brain magnetic resonance imaging. The participants with suspected cognitive impairment underwent additional examinations for detailed evaluation in the second phase. Statistical analysis was performed to identify risk and protective factors for dementia after all participants were diagnosed. RESULTS There were 927 participants in the baseline evaluation; 611 (65.9%) were healthy, 165 (17.8%) had mild cognitive impairment (MCI), and 151 (16.3%) had dementia. The age-standardised prevalence of dementia was 9.5%. Statistical analyses for amnestic MCI and Alzheimer's disease showed that risk factors for cognitive decline were diabetes mellitus, low activities of daily living, and living alone, and that protective factors were history of exercise and drinking habit. CONCLUSION The present findings suggest that several lifestyle-related diseases and factors are associated with cognitive decline. These results support similar findings from previous studies and will be helpful for preventing dementia in the future.
Collapse
Affiliation(s)
- Taku Yoshida
- Department of Psychiatry, Zaidan Niihama Hospital, Niihama, Japan
- Department of Neuropsychiatry, Ehime University Graduate School of Medicine, Toon, Japan
| | - Takaaki Mori
- Department of Neuropsychiatry, Ehime University Graduate School of Medicine, Toon, Japan
- Department of Psychiatry, Heisei Hospital, Ozu, Japan
| | - Hideaki Shimizu
- Department of Neuropsychiatry, Ehime University Graduate School of Medicine, Toon, Japan
- Department of Psychiatry, Heisei Hospital, Ozu, Japan
| | - Ayumi Tachibana
- Department of Neuropsychiatry, Ehime University Graduate School of Medicine, Toon, Japan
- Department of Psychiatry, Matsukaze Hospital, Shikokuchuou, Japan
| | - Yuta Yoshino
- Department of Neuropsychiatry, Ehime University Graduate School of Medicine, Toon, Japan
| | - Shinichiro Ochi
- Department of Neuropsychiatry, Ehime University Graduate School of Medicine, Toon, Japan
| | - Kiyohiro Yamazaki
- Department of Neuropsychiatry, Ehime University Graduate School of Medicine, Toon, Japan
| | - Yuki Ozaki
- Department of Neuropsychiatry, Ehime University Graduate School of Medicine, Toon, Japan
| | - Kentaro Kawabe
- Department of Neuropsychiatry, Ehime University Graduate School of Medicine, Toon, Japan
| | - Fumie Horiuchi
- Department of Neuropsychiatry, Ehime University Graduate School of Medicine, Toon, Japan
| | - Kenjiro Komori
- Department of Psychiatry, Zaidan Niihama Hospital, Niihama, Japan
- Department of Neuropsychiatry, Ehime University Graduate School of Medicine, Toon, Japan
- Office of Psychology, Department of Psychiatry, Juzen-Yurinoki Hospital, Niihama, Japan
| | - Jun-Ichi Iga
- Department of Neuropsychiatry, Ehime University Graduate School of Medicine, Toon, Japan
| | - Shu-Ichi Ueno
- Department of Neuropsychiatry, Ehime University Graduate School of Medicine, Toon, Japan
| |
Collapse
|
9
|
Wang W, Zhang L, Cao W, Xia K, Huo J, Huang T, Fan D. Systematic Screening of Associations between Medication Use and Risk of Neurodegenerative Diseases Using a Mendelian Randomization Approach. Biomedicines 2023; 11:1930. [PMID: 37509570 PMCID: PMC10377701 DOI: 10.3390/biomedicines11071930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 07/01/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND Systematically assessing the causal associations between medications and neurodegenerative diseases is significant in identifying disease etiology and novel therapies. Here, we investigated the putative causal associations between 23 existing medication categories and major neurodegenerative diseases (NDs), including Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). METHODS A two-sample mendelian randomization (MR) approach was conducted. Estimates were calculated using the inverse-variance weighted (IVW) method as the main model. A sensitivity analysis and a pleiotropy analysis were performed to identify potential violations. RESULTS Genetically predisposition to antihypertensives (OR = 0.809, 95% CI = 0.668-0.981, p = 0.031), thyroid preparations (OR = 0.948, 95% CI = 0.909-0.988, p = 0.011), and immunosuppressants (OR = 0.879, 95% CI = 0.789-0.979, p = 0.018) was associated with a decreased risk of AD. Genetic proxies for thyroid preparations (OR = 0.934, 95% CI = 0.884-0.988, p = 0.017), immunosuppressants (OR = 0.825, 95% CI = 0.699-0.973, p = 0.022), and glucocorticoids (OR = 0.862, 95% CI = 0.756-0.983, p = 0.027) were causally associated with a decreased risk of PD. Genetically determined antithrombotic agents (OR = 1.234, 95% CI = 1.042-1.461, p = 0.015), HMG CoA reductase inhibitors (OR = 1.085, 95% CI = 1.025-1.148, p = 0.005), and salicylic acid and derivatives (OR = 1.294, 95% CI = 1.078-1.553, p = 0.006) were associated with an increased risk of ALS. CONCLUSIONS We presented a systematic view concerning the causal associations between medications and NDs, which will promote the etiology discovery, drug repositioning and patient management for NDs.
Collapse
Affiliation(s)
- Wenjing Wang
- Department of Neurology, Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing 100191, China
- Key Laboratory for Neuroscience, National Health Commission/Ministry of Education, Peking University, Beijing 100191, China
| | - Linjing Zhang
- Department of Neurology, Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing 100191, China
- Key Laboratory for Neuroscience, National Health Commission/Ministry of Education, Peking University, Beijing 100191, China
| | - Wen Cao
- Department of Neurology, Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing 100191, China
- Key Laboratory for Neuroscience, National Health Commission/Ministry of Education, Peking University, Beijing 100191, China
| | - Kailin Xia
- Department of Neurology, Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing 100191, China
- Key Laboratory for Neuroscience, National Health Commission/Ministry of Education, Peking University, Beijing 100191, China
| | - Junyan Huo
- Department of Neurology, Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing 100191, China
- Key Laboratory for Neuroscience, National Health Commission/Ministry of Education, Peking University, Beijing 100191, China
| | - Tao Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing 100191, China
- Key Laboratory of Molecular Cardiovascular Sciences, Peking University, Ministry of Education, Beijing 100191, China
| | - Dongsheng Fan
- Department of Neurology, Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing 100191, China
- Key Laboratory for Neuroscience, National Health Commission/Ministry of Education, Peking University, Beijing 100191, China
| |
Collapse
|
10
|
Hammadi SH, Hassan MA, Allam EA, Elsharkawy AM, Shams SS. Effect of sacubitril/valsartan on cognitive impairment in colchicine-induced Alzheimer's model in rats. Fundam Clin Pharmacol 2023; 37:275-286. [PMID: 36203311 DOI: 10.1111/fcp.12837] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 08/19/2022] [Accepted: 10/05/2022] [Indexed: 11/05/2022]
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disease. There is epidemiological evidence that heart failure (HF) patients are at higher risk of developing AD, and the impact of sacubitril/valsartan, the first angiotensin receptor-neprilysin inhibitor (ARNI) approved for HF, on cognitive functions is still controversial. To investigate the effect of sacubitril/valsartan on cognitive functions in colchicine-induced AD rat model. Forty adult male Wistar rats were equally allocated into four groups (each of 10 rats): Group I: normal control, Group II: intracerebroventricular injection of colchicine (15 μg/5 μl/bilaterally), Group III: colchicine (15 μg/5 μl/bilaterally, icv) + oral sacubitril/valsartan (100 mg/kg/day) for 25 days, and Group IV: colchicine (15 μg/5 μl/bilaterally, icv) + oral valsartan (50 mg/kg/day) for 25 days. Behavioral assessment was done using Morris water maze and passive avoidance tasks. Biochemically, β-amyloid (1-40 and 1-42) peptides, oxidative stress (malondialdehyde and superoxide dismutase) and inflammatory (tumor necrosis factor-alpha) parameters were measured in hippocampus and prefrontal cortex. Sacubitril/valsartan exaggerated colchicine-induced cognitive impairment in both Morris water maze and passive avoidance tasks and was associated with significant increase in β-amyloid accumulation, oxidative stress, and inflammation versus valsartan. Sacubitril/valsartan caused deleterious effect on cognitive impairment and biochemical alterations in colchicine-induced AD rat model. Hence, special caution should be taken following long-term intake of ARNI on cognitive functions.
Collapse
Affiliation(s)
- Sami H Hammadi
- Clinical Pharmacology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Madiha A Hassan
- Clinical Pharmacology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Eman A Allam
- Medical Physiology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Amal M Elsharkawy
- Clinical Pharmacology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Sherouk S Shams
- Clinical Pharmacology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| |
Collapse
|
11
|
García-Lluch G, Peña-Bautista C, Royo LM, Baquero M, Cañada-Martínez AJ, Cháfer-Pericás C. Angiotensin II Receptor Blockers Reduce Tau/Aß42 Ratio: A Cerebrospinal Fluid Biomarkers’ Case-Control Study. Pharmaceutics 2023; 15:pharmaceutics15030924. [PMID: 36986785 PMCID: PMC10059654 DOI: 10.3390/pharmaceutics15030924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/03/2023] [Accepted: 03/03/2023] [Indexed: 03/16/2023] Open
Abstract
(1) Background: The role of antihypertensives in Alzheimer’s Disease (AD) prevention is controversial. This case-control study aims to assess whether antihypertensive medication has a protective role by studying its association with amyloid and tau abnormal levels. Furthermore, it suggests a holistic view of the involved pathways between renin-angiotensin drugs and the tau/amyloidß42 ratio (tau/Aß42 ratio); (2) Methods: The medical records of the participant patients were reviewed, with a focus on prescribed antihypertensive drugs and clinical variables, such as arterial blood pressure. The Anatomical Therapeutic Chemical classification was used to classify each drug. The patients were divided into two groups: patients with AD diagnosis (cases) and cognitively healthy patients (control); (3) Results: Age and high systolic blood pressure are associated with a higher risk of developing AD. In addition, combinations of angiotensin II receptor blockers are associated with a 30% lower t-tau/Aß42 ratio than plain angiotensin-converting enzyme inhibitor consumption; (4) Conclusions: Angiotensin II receptor blockers may play a potential role in neuroprotection and AD prevention. Likewise, several mechanisms, such as the PI3K/Akt/GSK3ß or the ACE1/AngII/AT1R axis, may link cardiovascular pathologies and AD presence, making its modulation a pivotal point in AD prevention. The present work highlights the central pathways in which antihypertensives may affect the presence of pathological amyloid and tau hyperphosphorylation.
Collapse
Affiliation(s)
- Gemma García-Lluch
- Research Group in Alzheimer Disease, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
- Cátedra DeCo MICOF-CEU UCH, Universidad Cardenal Herrera-CEU, 46115 Valencia, Spain
| | - Carmen Peña-Bautista
- Research Group in Alzheimer Disease, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
| | - Lucrecia Moreno Royo
- Cátedra DeCo MICOF-CEU UCH, Universidad Cardenal Herrera-CEU, 46115 Valencia, Spain
- Department of Pharmacy, Universidad Cardenal Herrera-CEU, CEU Universities, 46115 Valencia, Spain
| | - Miguel Baquero
- Research Group in Alzheimer Disease, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
- Cátedra DeCo MICOF-CEU UCH, Universidad Cardenal Herrera-CEU, 46115 Valencia, Spain
- Neurology Unit, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain
| | | | - Consuelo Cháfer-Pericás
- Research Group in Alzheimer Disease, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
- Cátedra DeCo MICOF-CEU UCH, Universidad Cardenal Herrera-CEU, 46115 Valencia, Spain
- Correspondence:
| |
Collapse
|
12
|
Kodam P, Sai Swaroop R, Pradhan SS, Sivaramakrishnan V, Vadrevu R. Integrated multi-omics analysis of Alzheimer's disease shows molecular signatures associated with disease progression and potential therapeutic targets. Sci Rep 2023; 13:3695. [PMID: 36879094 PMCID: PMC9986671 DOI: 10.1038/s41598-023-30892-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by the formation of amyloid plaques implicated in neuronal death. Genetics, age, and sex are the risk factors attributed to AD. Though omics studies have helped to identify pathways associated with AD, an integrated systems analysis with the available data could help to understand mechanisms, potential biomarkers, and therapeutic targets. Analysis of transcriptomic data sets from the GEO database, and proteomic and metabolomic data sets from literature was performed to identify deregulated pathways and commonality analysis identified overlapping pathways among the data sets. The deregulated pathways included those of neurotransmitter synapses, oxidative stress, inflammation, vitamins, complement, and coagulation pathways. Cell type analysis of GEO data sets showed microglia, endothelial, myeloid, and lymphoid cells are affected. Microglia are associated with inflammation and pruning of synapses with implications for memory and cognition. Analysis of the protein-cofactor network of B2, B6, and pantothenate shows metabolic pathways modulated by these vitamins which overlap with the deregulated pathways from the multi-omics analysis. Overall, the integrated analysis identified the molecular signature associated with AD. Treatment with anti-oxidants, B2, B6, and pantothenate in genetically susceptible individuals in the pre-symptomatic stage might help in better management of the disease.
Collapse
Affiliation(s)
- Pradeep Kodam
- Department of Biological Sciences, Birla Institute of Technology and Science Pilani, Hyderabad Campus, Jawahar Nagar, Hyderabad, Telangana, 500078, India
| | - R Sai Swaroop
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Prasanthi Nilayam, Anantapur, Andhra Pradesh, 515134, India
| | - Sai Sanwid Pradhan
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Prasanthi Nilayam, Anantapur, Andhra Pradesh, 515134, India
| | - Venketesh Sivaramakrishnan
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Prasanthi Nilayam, Anantapur, Andhra Pradesh, 515134, India.
| | - Ramakrishna Vadrevu
- Department of Biological Sciences, Birla Institute of Technology and Science Pilani, Hyderabad Campus, Jawahar Nagar, Hyderabad, Telangana, 500078, India.
| |
Collapse
|
13
|
Carey A, Fossati S. Hypertension and hyperhomocysteinemia as modifiable risk factors for Alzheimer's disease and dementia: New evidence, potential therapeutic strategies, and biomarkers. Alzheimers Dement 2023; 19:671-695. [PMID: 36401868 PMCID: PMC9931659 DOI: 10.1002/alz.12871] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/04/2022] [Accepted: 10/17/2022] [Indexed: 11/21/2022]
Abstract
This review summarizes recent evidence on how mid-life hypertension, hyperhomocysteinemia (HHcy) and blood pressure variability, as well as late-life hypotension, exacerbate Alzheimer's disease (AD) and dementia risk. Intriguingly, HHcy also increases the risk for hypertension, revealing the importance of understanding the relationship between comorbid cardiovascular risk factors. Hypertension-induced dementia presents more evidently in women, highlighting the relevance of sex differences in the impact of cardiovascular risk. We summarize each major antihypertensive drug class's effects on cognitive impairment and AD pathology, revealing how carbonic anhydrase inhibitors, diuretics modulating cerebral blood flow, have recently gained preclinical evidence as promising treatment against AD. We also report novel vascular biomarkers for AD and dementia risk, highlighting those associated with hypertension and HHcy. Importantly, we propose that future studies should consider hypertension and HHcy as potential contributors to cognitive impairment, and that uncovering the underlying molecular mechanisms and biomarkers would aid in the identification of preventive strategies.
Collapse
Affiliation(s)
- Ashley Carey
- Alzheimer’s Center at Temple, Department of Neural Sciences, Temple University Lewis Katz School of Medicine, Philadelphia
| | - Silvia Fossati
- Alzheimer’s Center at Temple, Department of Neural Sciences, Temple University Lewis Katz School of Medicine, Philadelphia
| |
Collapse
|
14
|
Kuber B, Fadnavis M, Chatterjee B. Role of angiotensin receptor blockers in the context of Alzheimer's disease. Fundam Clin Pharmacol 2023; 37:429-445. [PMID: 36654189 DOI: 10.1111/fcp.12872] [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: 06/14/2022] [Revised: 12/06/2022] [Accepted: 01/13/2023] [Indexed: 01/20/2023]
Abstract
As the world's population ages, the prevalence of age-related neurological disorders such as Alzheimer's disease (AD) is increasing. There is currently no treatment for Alzheimer's disease, and the few approved medications have a low success rate in lowering symptoms. As a result, several attempts are underway worldwide to identify new targets for the therapy of Alzheimer's disease. In preclinical studies of Alzheimer's disease, it was recently found that inhibition of angiotensin-converting enzyme (ACE) and blocking of the angiotensin II receptors reduce symptoms of neurodegeneration, Aβ plaque development, and tau hyperphosphorylation. Angiotensin II type I (AT1) blockers, such as telmisartan, candesartan, valsartan, and others, have a wide safety margin and are commonly used to treat hypertension. Renal and cardiovascular failures are reduced due to their vascular protective actions. Inhibition of AT1 receptors in the brain has a neuroprotective impact in humans, reducing the risk of stroke, increasing cognition, and slowing the progression of Alzheimer's disease. The review focuses on the mechanisms via which AT1 blockers may act beneficially in Alzheimer's disease. Although their effect is evident in preclinical studies, clinical trials, on the other hand, are in short supply to validate the strategy. More dose-response experiments with possible AT1 blockers and brain-targeted administration will be needed in the future.
Collapse
Affiliation(s)
- Binal Kuber
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's Narsee Monjee Institute of Management Studies, Mumbai, India
| | - Mitisha Fadnavis
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's Narsee Monjee Institute of Management Studies, Mumbai, India
| | - Bappaditya Chatterjee
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's Narsee Monjee Institute of Management Studies, Mumbai, India
| |
Collapse
|
15
|
Louise R, Manon L, Vincent E, Andréanne L, Philippe B, Cyntia T, Bennett DA, Sébastien H, Frédéric C. Higher Angiotensin I Converting Enzyme 2 (ACE2) levels in the brain of individuals with Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.17.524254. [PMID: 36711734 PMCID: PMC9882134 DOI: 10.1101/2023.01.17.524254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a major cause of death in the elderly. Cognitive decline due to Alzheimer's disease (AD) is frequent in the geriatric population disproportionately affected by the COVID-19 pandemic. Interestingly, central nervous system (CNS) manifestations have been reported in SARS-CoV-2-infected patients. In this study, we investigated the levels of Angiotensin I Converting Enzyme 2 (ACE2), the main entry receptor of SARS-COV-2 in cells, in postmortem parietal cortex samples from two independent AD cohorts, totalling 142 persons. Higher concentrations of ACE2 protein and mRNA were found in individuals with a neuropathological diagnosis of AD compared to age-matched healthy control subjects. Brain levels of soluble ACE2 were inversely associated with cognitive scores (p = 0.02), markers of pericytes (PDGFRβ, p=0.02 and ANPEP, p = 0.007) and caveolin1 (p = 0.03), but positively correlated with soluble amyloid-β peptides (Aβ) concentrations (p = 0.01) and insoluble phospho- tau (S396/404, p = 0.002). No significant differences in ACE2 were observed in the 3xTgAD mouse model of tau and Aβ neuropathology. Results from immunofluorescence and Western blots showed that ACE2 protein is mainly localized in neurons in the human brain but predominantly in microvessels in the mouse brain. The present data show that an AD diagnosis is associated with higher levels of soluble ACE2 in the human brain, which might contribute to a higher risk of CNS SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Reveret Louise
- Faculty of pharmacy, Laval University, Quebec, QC, Canada
- CHU de Quebec Research Center, Quebec, QC, Canada
| | - Leclerc Manon
- Faculty of pharmacy, Laval University, Quebec, QC, Canada
- CHU de Quebec Research Center, Quebec, QC, Canada
| | | | | | - Bourassa Philippe
- Faculty of pharmacy, Laval University, Quebec, QC, Canada
- CHU de Quebec Research Center, Quebec, QC, Canada
| | | | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Hébert Sébastien
- CHU de Quebec Research Center, Quebec, QC, Canada
- Faculty of medicine, Laval University, Quebec, QC, Canada
| | - Calon Frédéric
- Faculty of pharmacy, Laval University, Quebec, QC, Canada
- CHU de Quebec Research Center, Quebec, QC, Canada
| |
Collapse
|
16
|
Rukavina Mikusic NL, Gironacci MM. Mas receptor endocytosis and signaling in health and disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 194:49-65. [PMID: 36631200 DOI: 10.1016/bs.pmbts.2022.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The renin angiotensin system (RAS) plays a major role in blood pressure regulation and electrolyte homeostasis and is mainly composed by two axes mediating opposite effects. The pressor axis, constituted by angiotensin (Ang) II and the Ang II type 1 receptor (AT1R), exerts vasoconstrictor, proliferative, hypertensive, oxidative and pro-inflammatory actions, while the depressor/protective axis, represented by Ang-(1-7), its Mas receptor (MasR) and the Ang II type 2 receptor (AT2R), opposes the actions elicited by the pressor arm. The MasR belongs to the G protein-coupled receptor (GPCR) family. To avoid receptor overstimulation, GPCRs undergo internalization and trafficking into the cell after being stimulated. Then, the receptor may induce other signaling cascades or it may even interact with other receptors, generating distinct biological responses. Thus, control of a GPCR regarding space and time affects the specificity of the signals transduced by the receptor and the ultimate cellular response. The present chapter is focused on the signaling and trafficking pathways of MasR under physiological conditions and its participation in the pathogenesis of numerous brain diseases.
Collapse
Affiliation(s)
- Natalia L Rukavina Mikusic
- From Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Dpto. Química Biológica, IQUIFIB (UBA-CONICET), Buenos Aires, Argentina
| | - Mariela M Gironacci
- From Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Dpto. Química Biológica, IQUIFIB (UBA-CONICET), Buenos Aires, Argentina.
| |
Collapse
|
17
|
El-Din Hussein AS, Abou-El Nour RKED, Khorshid OA, Osman AS. Study of the possible effect of sacubitril/valsartan combination versus valsartan on the cognitive function in Alzheimer's disease model in rats. Int J Immunopathol Pharmacol 2023; 37:3946320231161469. [PMID: 36877667 PMCID: PMC9996744 DOI: 10.1177/03946320231161469] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023] Open
Abstract
Objectives: Alzheimer's disease (AD) is an irreversible, progressive neurodegenerative disorder. The proportion of elderly individuals at risk for AD and cardiovascular problems increases by raising life expectancy. The present study was designed to investigate the effect of the sacubitril/valsartan combination compared to that of valsartan alone in a rat model of AD. Methods: 72 male adult Wistar rats were divided into seven groups; control untreated rats received saline, control valsartan-treated rats received valsartan orally, control sacubitril/valsartan treated rats received sacubitril/valsartan orally, model rats received aluminum chloride i.p., model valsartan treated rats received aluminum chloride i.p. and valsartan orally and model sacubitril/valsartan treated rats received aluminum chloride i.p. and sacubitril/valsartan combination orally. All previous treatments continued on a daily basis for 6 weeks. At the second, fourth, and sixth weeks of the experiment, behavioral changes were evaluated using the Morris water maze and novel object recognition tests, and systolic blood pressure was measured. In the end, rat brain malondialdehyde and amyloid-beta 1-42 levels were measured, and the isolated hippocampus was evaluated histopathologically. Results: Valsartan improved AD symptoms in the aluminum-induced rat model, while the sacubitril/valsartan combination significantly worsened all tested parameters in both control and model rats compared with untreated and valsartan-treated animals. Conclusion: Based on the current study's findings, valsartan did not increase the risk for AD development in control rats and improved AD symptoms in a rat model, while sacubitril/valsartan combination increased the risk of AD in control rats and worsened the condition in a rat model.
Collapse
Affiliation(s)
| | | | - Omayma A Khorshid
- Department of Medical Pharmacology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Afaf S Osman
- Department of Medical Pharmacology, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
18
|
Lee HW, Kim S, Jo Y, Kim Y, Ye BS, Yu YM. Neuroprotective effect of angiotensin II receptor blockers on the risk of incident Alzheimer's disease: A nationwide population-based cohort study. Front Aging Neurosci 2023; 15:1137197. [PMID: 36949774 PMCID: PMC10025478 DOI: 10.3389/fnagi.2023.1137197] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 02/13/2023] [Indexed: 03/08/2023] Open
Abstract
Background Recent studies on renin-angiotensin system (RAS) inhibitors have reported a reduced risk of Alzheimer's disease (AD). Nevertheless, the effect of RAS inhibitor type and blood-brain barrier (BBB) permeability on the risk of AD is still unknown. Objectives To assess the effects of RAS inhibitors on the risk of AD based on the type and BBB permeability and investigate the cumulative duration-response relationship. Methods This was a population-based retrospective cohort study using the Korean Health Insurance Review and Assessment database records from 2008 to 2019. The data of patients diagnosed with ischemic heart disease between January 2009 and June 2009 were identified for inclusion in the analyses. Propensity score matching was used to balance RAS inhibitor users with non-users. The association between the use of RAS inhibitors and incident AD was evaluated using a multivariate Cox proportional hazard regression model. The results are presented in adjusted hazard ratios (aHRs) and 95% confidence intervals (CIs). Results Among the 57,420 matched individuals, 7,303 developed AD within the follow-up period. While the use of angiotensin-converting enzyme inhibitors (ACEIs) was not significantly associated with AD risk, the use of angiotensin II receptor blockers (ARBs) showed a significant association with reduced risk of incident AD (aHR = 0.94; 95% CI = 0.90-0.99). Furthermore, the use of BBB-crossing ARBs was associated with a lower risk of AD (aHR = 0.83; 95% CI = 0.78-0.88) with a cumulative duration-response relationship. A higher cumulative dose or duration of BBB-crossing ARBs was associated with a gradual decrease in AD risk (P for trend < 0.001). No significant association between the use of ACEIs and the risk of AD was observed regardless of BBB permeability. Conclusion Long-term use of BBB-crossing ARBs significantly reduced the risk of AD development. The finding may provide valuable insight into disease-modifying drug options for preventing AD in patients with cardiovascular diseases.
Collapse
Affiliation(s)
- Hyun Woo Lee
- Department of Pharmaceutical Medicine and Regulatory Sciences, Colleges of Medicine and Pharmacy, Yonsei University, Incheon, Republic of Korea
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, Republic of Korea
| | - Seungyeon Kim
- College of Pharmacy, Dankook University, Cheonan, Republic of Korea
| | - Youngkwon Jo
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, Republic of Korea
| | - Youjin Kim
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, Republic of Korea
| | - Byoung Seok Ye
- Department of Neurology, College of Medicine, Yonsei University, Seoul, Republic of Korea
| | - Yun Mi Yu
- Department of Pharmaceutical Medicine and Regulatory Sciences, Colleges of Medicine and Pharmacy, Yonsei University, Incheon, Republic of Korea
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, Republic of Korea
- *Correspondence: Yun Mi Yu,
| |
Collapse
|
19
|
Kopp W. Pathogenesis of (smoking-related) non-communicable diseases-Evidence for a common underlying pathophysiological pattern. Front Physiol 2022; 13:1037750. [PMID: 36589440 PMCID: PMC9798240 DOI: 10.3389/fphys.2022.1037750] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
Non-communicable diseases, like diabetes, cardiovascular diseases, cancer, stroke, chronic obstructive pulmonary disease, osteoporosis, arthritis, Alzheimer's disease and other more are a leading cause of death in almost all countries. Lifestyle factors, especially poor diet and tobacco consumption, are considered to be the most important influencing factors in the development of these diseases. The Western diet has been shown to cause a significant distortion of normal physiology, characterized by dysregulation of the sympathetic nervous system, renin-angiotensin aldosterone system, and immune system, as well as disruption of physiological insulin and oxidant/antioxidant homeostasis, all of which play critical roles in the development of these diseases. This paper addresses the question of whether the development of smoking-related non-communicable diseases follows the same pathophysiological pattern. The evidence presented shows that exposure to cigarette smoke and/or nicotine causes the same complex dysregulation of physiology as described above, it further shows that the factors involved are strongly interrelated, and that all of these factors play a key role in the development of a broad spectrum of smoking-related diseases. Since not all smokers develop one or more of these diseases, it is proposed that this disruption of normal physiological balance represents a kind of pathogenetic "basic toolkit" for the potential development of a range of non-communicable diseases, and that the decision of whether and what disease will develop in an individual is determined by other, individual factors ("determinants"), such as the genome, epigenome, exposome, microbiome, and others. The common pathophysiological pattern underlying these diseases may provide an explanation for the often poorly understood links between non-communicable diseases and disease comorbidities. The proposed pathophysiological process offers new insights into the development of non-communicable diseases and may influence the direction of future research in both prevention and therapy.
Collapse
|
20
|
De Dios L, Collazo C, Inostroza-Nieves Y. Renin-angiotensin-system increases phosphorylated tau and Reactive Oxygen Species in human cortical neuron cell line. Biochem Biophys Rep 2022; 32:101355. [PMID: 36164564 PMCID: PMC9507985 DOI: 10.1016/j.bbrep.2022.101355] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 09/16/2022] [Accepted: 09/19/2022] [Indexed: 11/24/2022] Open
Abstract
Alzheimer's Disease (AD) is the most common cause of dementia. AD patients had increased extracellular amyloid β plaques and intracellular hyperphosphorylated tau (p-tau) in neurons. Recent studies have shown an association between the Renin-Angiotensin System (RAS) and AD. The involvement of RAS has been mediated through Angiotensin II (AngII), which is overexpressed in aging brains. However, the exact mechanism of how AngII contributes to AD is unknown. Thus, we hypothesize that AngII increases p-tau by activating its kinases, CDK5 and MAPK. In the human cortical neuron cell line, HCN2, treatment with AngII upregulated the gene expression of CDK5 (2.9 folds, p < 0.0001) and MAPTK (1.9 folds, p < 0.001). The AT1R antagonist, Losartan, blocked the changes in tau kinases. Also, AngII-induced the MAPK activation, increasing its phosphorylation by 400% (p < 0.0001), an increase that was also blocked by Losartan. An increase in p-tau by AngII was observed using fluorescent microscopy. We then quantified Reactive Oxygen Species (ROS) production, and it was significantly increased by AngII (p < 0.01), and treatment with Losartan blunted their production (p < 0.05). The data obtained demonstrated that AngII might contribute to the pathogenesis of AD. Angiotensin II increases CDK5 and MAPK gene expression in human cortical neuron cell line. Angiotensin II increases tau phosphorylation in human cortical neuron cell line. Angiotensin II increases Reactive Oxygen Species production in human cortical neuron cell line.
Collapse
|
21
|
Yang J, Gao Y, Duan Q, Qiu Y, Feng S, Zhan C, Huang Y, Zhang Y, Ma G, Nie K, Wang L. Renin-angiotensin system blockers affect cognitive decline in Parkinson's disease: The PPMI dataset. Parkinsonism Relat Disord 2022; 105:90-95. [PMID: 36395543 DOI: 10.1016/j.parkreldis.2022.10.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 10/13/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVE To explore the potential clinical effects of renin-angiotensin system blocker (RASB, angiotensin II receptor blockers (ARBs) and angiotensin-converting enzyme inhibitors (ACEIs)) in patients from the Parkinson's Progress Marker Initiative (PPMI) study database. METHODS One hundred and seven untreated, newly diagnosed PD patients with hypertension, from the PPMI were included. We measured cognitive performance, biomarkers in CSF, and magnetic resonance imaging (MRI) during the five follow-up years for patients exposed or not to renal-angiotensin system blockers. Sixteen PD patients with hypertension underwent [18F]florbetaben positron emission tomography (PET) scanning. SUVRs of region of interest (ROI) were calculated and compared within different groups. RESULT Treatment with ARBs but not ACEIs improved global cognitive function evaluated by MoCA score in PD patients with hypertension compared to other hypertensive medicines up to 5 years follow up. Specifically, ARBs improved visuospatial, memory, executive abilities, processing speed attention test scores in PD. There was no significant impact of ARBs on α-syn, tau, Aβ in CSF. RASBs reduced [18F] florbetaben uptake in cortex and subcortex nuclei in the brain. CONCLUSIONS These results show potential protective effect with ARBs in cognitive impairment of parkinson's disease with hypertension.
Collapse
Affiliation(s)
- Jianhua Yang
- The Second School of Clinical Medicine, Southern Medical University, No.1023, South Shatai Road, Baiyun District, Guangzhou, 510515, China; Department of Neurology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Neuroscience Institute, No.106, Zhongshan 2nd Road, Yuexiu District, Guangzhou, 510080, China; Department of Neurology, Ganzhou People's Hospital, Ganzhou, 341000, China
| | - Yuyuan Gao
- Department of Neurology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Neuroscience Institute, No.106, Zhongshan 2nd Road, Yuexiu District, Guangzhou, 510080, China
| | - Qingrui Duan
- Department of Neurology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Neuroscience Institute, No.106, Zhongshan 2nd Road, Yuexiu District, Guangzhou, 510080, China
| | - Yihui Qiu
- Department of Neurology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Neuroscience Institute, No.106, Zhongshan 2nd Road, Yuexiu District, Guangzhou, 510080, China
| | - Shujun Feng
- Department of Neurology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Neuroscience Institute, No.106, Zhongshan 2nd Road, Yuexiu District, Guangzhou, 510080, China
| | - Cuijing Zhan
- The Second School of Clinical Medicine, Southern Medical University, No.1023, South Shatai Road, Baiyun District, Guangzhou, 510515, China; Department of Neurology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Neuroscience Institute, No.106, Zhongshan 2nd Road, Yuexiu District, Guangzhou, 510080, China
| | - Yin Huang
- The Second School of Clinical Medicine, Southern Medical University, No.1023, South Shatai Road, Baiyun District, Guangzhou, 510515, China; Department of Neurology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Neuroscience Institute, No.106, Zhongshan 2nd Road, Yuexiu District, Guangzhou, 510080, China
| | - Yuhu Zhang
- Department of Neurology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Neuroscience Institute, No.106, Zhongshan 2nd Road, Yuexiu District, Guangzhou, 510080, China
| | - Guixian Ma
- Department of Neurology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Neuroscience Institute, No.106, Zhongshan 2nd Road, Yuexiu District, Guangzhou, 510080, China
| | - Kun Nie
- Department of Neurology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Neuroscience Institute, No.106, Zhongshan 2nd Road, Yuexiu District, Guangzhou, 510080, China.
| | - Lijuan Wang
- The Second School of Clinical Medicine, Southern Medical University, No.1023, South Shatai Road, Baiyun District, Guangzhou, 510515, China; Department of Neurology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong Neuroscience Institute, No.106, Zhongshan 2nd Road, Yuexiu District, Guangzhou, 510080, China.
| |
Collapse
|
22
|
Malone JE, Elkasaby MI, Lerner AJ. Effects of Hypertension on Alzheimer's Disease and Related Disorders. Curr Hypertens Rep 2022; 24:615-625. [PMID: 36125695 DOI: 10.1007/s11906-022-01221-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/26/2022] [Indexed: 01/31/2023]
Abstract
PURPOSE OF REVIEW To review the pathophysiology of hypertension in Alzheimer's disease and related dementias and explore the current landscape of clinical trials involving treatment of hypertension to improve cognition. RECENT FINDINGS Hypertension is increasingly recognized as a contributor to cognitive impairment. Clinical trials that explore blood pressure reductions with cognitive outcomes have been promising. Various antihypertensives have been evaluated in clinical trials, with growing interest in those agents that impact the renin-angiotensin-aldosterone system due to its own association with cognitive impairment. No antihypertensive agent has been found to be superior to others in reducing cognitive impairment risk or conferring neuroprotective benefits. In this review, the pathophysiology of and clinical trial data involving hypertension and dementia will be explored. Hypertension is a significant risk factor for the development of neurodegenerative dementias, and clinical trials have been overall favorable in improving cognition by reductions in blood pressure using antihypertensive agents.
Collapse
Affiliation(s)
- Joseph E Malone
- Department of Neurology, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Mohamed I Elkasaby
- Department of Neurology, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Alan J Lerner
- Department of Neurology, University Hospitals Cleveland Medical Center, Cleveland, OH, USA.
- Case Western Reserve University School of Medicine, Cleveland, OH, USA.
| |
Collapse
|
23
|
Yasar S. Candesartan-the next anti-amyloid drug? Brain Commun 2022; 4:fcac293. [PMID: 36440098 PMCID: PMC9683388 DOI: 10.1093/braincomms/fcac293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 08/24/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022] Open
Abstract
This scientific commentary refers to 'Safety and biomarker effects of candesartan in non-hypertensive adults with prodromal Alzheimer's disease' by Hajjar et al. (https://doi.org/10.1093/braincomms/fcac270).
Collapse
Affiliation(s)
- Sevil Yasar
- Division of Geriatric Medicine and Gerontology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
24
|
Wang Y, Yuan P, Ding L, Zhu J, Qi X, Zhang Y, Li Y, Xia X, Zheng JC. Circulating extracellular vesicle-containing microRNAs reveal potential pathogenesis of Alzheimer’s disease. Front Cell Neurosci 2022; 16:955511. [PMID: 36339820 PMCID: PMC9630335 DOI: 10.3389/fncel.2022.955511] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 09/27/2022] [Indexed: 11/24/2022] Open
Abstract
The pathogenesis of Alzheimer’s disease (AD) remains unknown till today, hindering the research and development of AD therapeutics and diagnostics. Circulating extracellular vesicles (EVs) can be utilized as a new window to spy upon AD pathogenesis. Altered microRNA profiles were noted in both the cerebrospinal fluid (CSF)- and blood-isolated EVs of AD patients, implying the outstanding potential of circulating EV-containing miRNAs (CEmiRs) to serve as important regulators in AD pathogenesis. Although several CEmiRs were found to play a part in AD, the association of globally altered miRNA profiles in patients’ serum-derived EVs with AD pathogenesis remains unclear. In this study, we first investigated the miRNA profile in serum-derived EVs from AD, mild cognitive impairment (MCI) patients, and healthy individuals. We observed differential expression patterns of CEmiRs and classified them into 10 clusters. We identified the predicted targets of these differentially expressed CEmiRs (DECEmiRs) and analyzed their biological functions and interactions. Our study revealed the temporal regulation of complex and precise signaling networks on AD pathogenesis, shedding light on the development of novel therapeutic strategies, including multi-target drug combination for AD treatment.
Collapse
Affiliation(s)
- Yi Wang
- Translational Research Center, Shanghai Yangzhi Rehabilitation Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Ping Yuan
- Department of Cardio-Pulmonary Circulation, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Lu Ding
- Center for Translational Neurodegeneration and Regenerative Therapy, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Jie Zhu
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People’s Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Xinrui Qi
- Center for Translational Neurodegeneration and Regenerative Therapy, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Yanyan Zhang
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People’s Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Yunxia Li
- Department of Neurology, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
- Yunxia Li,
| | - Xiaohuan Xia
- Center for Translational Neurodegeneration and Regenerative Therapy, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People’s Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
- Shanghai Frontiers Science Center of Nanocatalytic Medicine, Tongji University, Shanghai, China
- Xiaohuan Xia,
| | - Jialin C. Zheng
- Center for Translational Neurodegeneration and Regenerative Therapy, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People’s Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
- Shanghai Frontiers Science Center of Nanocatalytic Medicine, Tongji University, Shanghai, China
- Collaborative Innovation Center for Brain Science, Tongji University, Shanghai, China
- *Correspondence: Jialin C. Zheng,
| |
Collapse
|
25
|
Ağagündüz D, Gençer Bingöl F, Çelik E, Cemali Ö, Özenir Ç, Özoğul F, Capasso R. Recent developments in the probiotics as live biotherapeutic products (LBPs) as modulators of gut brain axis related neurological conditions. Lab Invest 2022; 20:460. [PMID: 36209124 PMCID: PMC9548122 DOI: 10.1186/s12967-022-03609-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 08/24/2022] [Indexed: 11/10/2022]
Abstract
Probiotics have been defined as “living microorganisms that create health benefits in the host when taken in sufficient amounts. Recent developments in the understanding of the relationship between the microbiom and its host have shown evidence about the promising potential of probiotics to improve certain health problems. However, today, there are some confusions about traditional and new generation foods containing probiotics, naming and classifications of them in scientific studies and also their marketing. To clarify this confusion, the Food and Drug Administration (FDA) declared that it has made a new category definition called "live biotherapeutic products" (LBPs). Accordingly, the FDA has designated LBPs as “a biological product that: i)contains live organisms, such as bacteria; ii)is applicable to the prevention, treatment, or cure of a disease/condition of human beings; and iii) is not a vaccine”. The accumulated literature focused on LBPs to determine effective strains in health and disease, and often focused on obesity, diabetes, and certain diseases like inflammatory bowel disease (IBD).However, microbiome also play an important role in the pathogenesis of diseases that age day by day in the modern world via gut-brain axis. Herein, we discuss the novel roles of LBPs in some gut-brain axis related conditions in the light of recent studies. This article may be of interest to a broad readership including those interested in probiotics as LBPs, their health effects and safety, also gut-brain axis.
Collapse
Affiliation(s)
- Duygu Ağagündüz
- Department of Nutrition and Dietetics, Gazi University, Faculty of Health Sciences, 06490, Ankara, Emek, Turkey.
| | - Feray Gençer Bingöl
- Department of Nutrition and Dietetics, Burdur Mehmet Akif Ersoy University, İstiklal Yerleşkesi, 15030, Burdur, Turkey
| | - Elif Çelik
- Department of Nutrition and Dietetics, Gazi University, Faculty of Health Sciences, 06490, Ankara, Emek, Turkey
| | - Özge Cemali
- Department of Nutrition and Dietetics, Gazi University, Faculty of Health Sciences, 06490, Ankara, Emek, Turkey
| | - Çiler Özenir
- Department of Nutrition and Dietetics, Kırıkkale University, 71100, Kırıkkale, Merkez, Turkey
| | - Fatih Özoğul
- Department of Seafood Processing Technology, Faculty of Fisheries, Cukurova University, 01330, Balcali, Adana, Turkey
| | - Raffaele Capasso
- Department of Agricultural Sciences, University of Naples Federico II, 80055, Portici, NA, Italy.
| |
Collapse
|
26
|
Antidepressant-like Effects of Renin Inhibitor Aliskiren in an Inflammatory Mouse Model of Depression. Brain Sci 2022; 12:brainsci12050655. [PMID: 35625041 PMCID: PMC9139539 DOI: 10.3390/brainsci12050655] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/09/2022] [Accepted: 05/13/2022] [Indexed: 11/16/2022] Open
Abstract
Depression is considered a neuropsychic disease that has global prevalence and is associated with disability. The pathophysiology of depression is not well understood; however, emerging evidence has indicated that neuroinflammation could contribute to developing depression symptoms. One of the factors that have a role in the development of neuroinflammation is the renin–angiotensin system. Therefore, the goal of the current study is to determine the antidepressant-like effects of Aliskiren, a renin inhibitor, against lipopolysaccharide (LPS)-induced depressive-like behavior in mice, glial cell activation, and the upregulation of proinflammatory cytokines in the prefrontal cortex. For behavioral studies, the open field test (OFT), tail suspension test (TST), forced swim test (FST), and sucrose preference test (SPT) were used. Inflammatory markers were assessed using real-time polymerase chain reaction (RT-PCR). LPS administration (0.5 mg/kg, intraperitoneal injection (i.p.)) sufficiently reduced the number of crossings in OFT, whereas Aliskiren pretreatment (10 mg/kg, i.p.) attenuated the LPS effect for two hours after LPS injection. The treatments did not show effects on locomotor activity in OFT 24 h after LPS administration. LPS increased the immobility time in TST and FST or reduced sucrose consumption in SPT after 24 h. Aliskiren reversed the effects induced by LPS in TST, FST, and SPT. CD11 b mRNA, a microglial marker, GFAP mRNA, an astroglial marker, and proinflammatory cytokines genes (TNF-α, IL-1β, and IL-6) were upregulated in the prefrontal cortex in LPS exposed animals. However, Aliskiren reduced LPS-induced inflammatory genes in the prefrontal cortex. Hence, the outcomes conclude that Aliskiren prevents depressive illness associated with neuroinflammation in humans.
Collapse
|
27
|
Cosarderelioglu C, Nidadavolu LS, George CJ, Marx-Rattner R, Powell L, Xue QL, Tian J, Salib J, Oh ES, Ferrucci L, Dincer P, Bennett DA, Walston JD, Abadir PM. Higher Angiotensin II Type 1 Receptor Levels and Activity in the Postmortem Brains of Older Persons with Alzheimer's Dementia. J Gerontol A Biol Sci Med Sci 2022; 77:664-672. [PMID: 34914835 PMCID: PMC8974324 DOI: 10.1093/gerona/glab376] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Indexed: 11/12/2022] Open
Abstract
Aging is a key risk factor in Alzheimer's dementia (AD) development and progression. The primary dementia-protective benefits of angiotensin II subtype 1 receptor (AT1R) blockers are believed to arise from systemic effects on blood pressure. However, a brain-specific renin-angiotensin system (b-RAS) exists, which can be altered by AT1R blockers. Brain RAS acts mainly through 3 angiotensin receptors: AT1R, AT2R, and AT4R. Changes in these brain angiotensin receptors may accelerate the progression of AD. Using postmortem frontal cortex brain samples of age- and sex-matched cognitively normal individuals (n = 30) and AD patients (n = 30), we sought to dissect the b-RAS changes associated with AD and assess how these changes correlate with brain markers of oxidative stress, inflammation, and mitochondrial dysfunction as well as amyloid-β and paired helical filament tau pathologies. Our results show higher protein levels of the pro-inflammatory AT1R and phospho-ERK (pERK) in the brains of AD participants. Brain AT1R levels and pERK correlated with higher oxidative stress, lower cognitive performance, and higher tangle and amyloid-β scores. This study identifies molecular changes in b-RAS and offers insight into the role of b-RAS in AD-related brain pathology.
Collapse
Affiliation(s)
- Caglar Cosarderelioglu
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Internal Medicine, Division of Geriatrics, Ankara University School of Medicine, Ankara, Turkey
- Department of Medical Biology, Hacettepe University School of Medicine, Ankara, Turkey
| | - Lolita S Nidadavolu
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Claudene J George
- Department of Medicine, Division of Geriatrics, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, New York, USA
| | - Ruth Marx-Rattner
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Laura Powell
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Qian-Li Xue
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Johns Hopkins University Center on Aging and Health, Baltimore, Maryland, USA
| | - Jing Tian
- Department of Biostatistics, Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Joy Salib
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Esther S Oh
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Luigi Ferrucci
- National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Pervin Dincer
- Department of Medical Biology, Hacettepe University School of Medicine, Ankara, Turkey
| | - David A Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, Illinois, USA
| | - Jeremy D Walston
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Peter M Abadir
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
28
|
Jo Y, Kim S, Ye BS, Lee E, Yu YM. Protective Effect of Renin-Angiotensin System Inhibitors on Parkinson's Disease: A Nationwide Cohort Study. Front Pharmacol 2022; 13:837890. [PMID: 35308220 PMCID: PMC8927987 DOI: 10.3389/fphar.2022.837890] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 02/14/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Renin-angiotensin system (RAS) inhibitors have been suggested as protective agents in Parkinson's disease (PD). However, epidemiological evidence on the association between RAS inhibitors and the development of PD is inconsistent. Objectives: To investigate the effect of RAS inhibitors on PD risk in patients with ischemic heart disease (IHD) by type and cumulative duration of RAS inhibitors and their degree of blood-brain barrier (BBB) penetration ability. Methods: This was a propensity score-matched retrospective cohort study using 2008-2019 healthcare claims data from the Korean Health Insurance Review and Assessment database. The association between RAS inhibitor use and PD in patients with IHD was evaluated using multivariate Cox proportional hazard regression analysis. The risks are presented as adjusted hazard ratios (aHRs) and 95% confidence intervals (CIs). Results: Over a 10-year follow-up, 1,086 of 62,228 IHD patients developed PD. The Cox regression model showed that the use of RAS inhibitors was significantly associated with a lower risk of PD (aHR = 0.75; 95% CI 0.66-0.85) than the non-use of RAS inhibitors. Specifically, this reduced risk of PD only remained with the use of BBB-crossing angiotensin II receptor blockers (ARBs) (aHR = 0.62; 95% CI = 0.53-0.74), and this association was more definite with an increasing cumulative duration. A significantly reduced risk of PD was not observed with the use of BBB-crossing angiotensin-converting enzyme inhibitors. Conclusions: The use of ARBs with BBB-penetrating properties and a high cumulative duration significantly reduces the risk of PD in IHD patients. This protective effect could provide insight into disease-modifying drug candidates for PD.
Collapse
Affiliation(s)
- Youngkwon Jo
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, South Korea
| | - Seungyeon Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea.,Transdisciplinary Department of Medicine and Advanced Technology, Seoul National University Hospital, Seoul, South Korea
| | - Byoung Seok Ye
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea
| | - Euni Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea
| | - Yun Mi Yu
- Department of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, South Korea.,Department of Pharmaceutical Medicine and Regulatory Sciences, Colleges of Medicine and Pharmacy, Yonsei University, Incheon, South Korea
| |
Collapse
|
29
|
Sood A, Goyal R, Singh H, Behl T, Arora S, Saini B, Kaur R. Implication of Covid-19 on Neurological Complications with Specific Emphasis on Alzheimer's and Parkinson's Disease. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2022; 21:235-245. [PMID: 34414876 DOI: 10.2174/1871527320666210820092817] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 05/02/2021] [Accepted: 06/25/2021] [Indexed: 01/10/2023]
Abstract
It is noticeable how the novel coronavirus has spread from the Wuhan region of China to the whole world, devastating the lives of people worldwide. All the data related to the precautionary measures, diagnosis, treatment, and even the epidemiological data are being made freely accessible and reachable in a very little time as well as being rapidly published to save humankind from this pandemic. There might be neurological complications of COVID-19 and patients suffering from neurodegenerative conditions like Alzheimer's disease and Parkinson's disease might have repercussions as a result of the pandemic. In this review article, we have discussed the effect of SARS-CoV-2 viral infection on the people affected with neurodegenerative disorders such as Parkinson's and Alzheimer's. It primarily emphasizes two issues, i.e., vulnerability to infection and modifications of course of the disease concerning the clinical neurological manifestations, the advancement of the disease and novel approaches to support health care professionals in disease management, the susceptibility to these diseases, and impact on the severity of disease and management.
Collapse
Affiliation(s)
- Ankita Sood
- Chitkara College of Pharmacy, Chitkara University, Punjab,India
| | - Ravi Goyal
- Chitkara College of Pharmacy, Chitkara University, Punjab,India
| | - Harshdeep Singh
- Chitkara College of Applied Engineering, Chitkara University, Punjab,India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab,India
| | - Sandeep Arora
- Chitkara College of Pharmacy, Chitkara University, Punjab,India
| | - Balraj Saini
- Chitkara College of Pharmacy, Chitkara University, Punjab,India
| | - Rajwinder Kaur
- Chitkara College of Pharmacy, Chitkara University, Punjab,India
| |
Collapse
|
30
|
Mohapatra D, Kanungo S, Pradhan SP, Jena S, Prusty SK, Sahu PK. Captopril is more effective than Perindopril against aluminium chloride induced amyloidogenesis and AD like pathology. Heliyon 2022; 8:e08935. [PMID: 35243060 PMCID: PMC8857426 DOI: 10.1016/j.heliyon.2022.e08935] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/04/2021] [Accepted: 02/08/2022] [Indexed: 11/15/2022] Open
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disorder. Aluminium chloride induces AD like pathology in rats. Renin angiotensin system plays a significant role in the pathogenesis and occurrence of Alzheimer's disease. In the present study we evaluated and compared the effect of Captopril and Perindopril against aluminium chloride induced amyloidogenesis and cognitive dysfunction in rats. Wistar rats of both sex were divided randomly into four groups i.e. Group I was served as normal control and treated with normal saline, Group II was administered with AlCl3 (100 mg/kg, p. o.) and Group III and IV received Captopril (30 mg/kg, p. o.) and Perindopril (5 mg/kg, p. o.) respectively 1hr prior to administration of AlCl3. All the doses were given once daily for 42 days. The evaluation of memory function was carried out in Y-maze (spontaneous alternation), radial arm maze (number of correct responses) and elevated plus maze (transfer latency). After behavioral studies, estimation of antioxidant status (brain and serum), amyloid-β content (brain) and histopathology of brain hippocampus region was done. Administration of AlCl3 for 42 days impaired cognitive dysfunction. Captopril and Perindopril prevented AlCl3 induced cognitive dysfunction by improving spontaneous alternation behavior, number of correct responses and reducing transfer latency. They also increase the antioxidant status, reduce the Aβ42 content in the brain and reverse the histopathological changes caused by AlCl3 in hippocampal region. Both Captopril and Perindopril protects against aluminium chloride induced amyloidogenesis and AD like pathology. Captopril is found to be more effective than Perindopril.
Collapse
Affiliation(s)
| | | | | | - Susmita Jena
- School of Pharmaceutical Sciences, Siksha O Anusandhan University, India
| | | | - Pratap Kumar Sahu
- School of Pharmaceutical Sciences, Siksha O Anusandhan University, India
| |
Collapse
|
31
|
Chen G, Zhou S, Deng F. Influence of Renin-Angiotensin System Inhibitors on Postoperative Delirium in Patients With Pulmonary Arterial Hypertension: A Secondary Analysis of a Retrospective Cohort Study. Front Psychiatry 2022; 13:851104. [PMID: 35463531 PMCID: PMC9024170 DOI: 10.3389/fpsyt.2022.851104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 03/08/2022] [Indexed: 12/04/2022] Open
Abstract
OBJECTIVE To investigate the correlation between preoperative use of ACEIs/ARBs and postoperative delirium (POD) in surgical patients with pulmonary arterial hypertension (PAH). METHODS The present study is a secondary analysis of a retrospective cohort study conducted at the University of Washington Medical Center from April 2007 to September 2013. Patients with PAH who underwent non-cardiac, non-obstetric surgery were enrolled in the original research. We further excluded stroke, sepsis, and craniotomy patients from interfering with POD evaluation. The univariate regression analysis and multivariate-adjusted model were used to explore the influence of preoperative ACEIs/ARBs use on the occurrence of POD. RESULTS A total of 539 patients were included in this study. The incidence of POD in these patients was 3.0%. Following the adjustment of potential confounders (age, BMI, smoking status, pulmonary arterial systolic pressure, length of surgery, vascular surgery, asthma, obstructive sleep apnea, renal failure, atrial fibrillation, coronary artery disease, hydrochlorothiazide, alpha-blocker, calcium channel blocker, antiplatelet, steroids, statin, isoflurane), a negative relationship was found between preoperative use of ACEIs/ARBs and occurrence of POD (OR = 0.15, 95%CI: 0.03 to 0.80, P = 0.0266). CONCLUSION Preoperative use of ACEIs/ARBs in patients with PAH reduces the risk of POD. ACEIs/ARBs may be more recommended for patients with PAH in the future.
Collapse
Affiliation(s)
- Gong Chen
- Department of Anesthesiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Sai Zhou
- Academic Affairs Department, Changsha Medical University, Changsha, China
| | - Fang Deng
- Department of Anesthesiology, Xiangya Changde Hospital, Changde, China
| |
Collapse
|
32
|
Le D, Brown L, Malik K, Murakami S. Two Opposing Functions of Angiotensin-Converting Enzyme (ACE) That Links Hypertension, Dementia, and Aging. Int J Mol Sci 2021; 22:ijms222413178. [PMID: 34947975 PMCID: PMC8707689 DOI: 10.3390/ijms222413178] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/01/2021] [Accepted: 12/05/2021] [Indexed: 01/18/2023] Open
Abstract
A 2018 report from the American Heart Association shows that over 103 million American adults have hypertension. The angiotensin-converting enzyme (ACE) (EC 3.4.15.1) is a dipeptidyl carboxylase that, when inhibited, can reduce blood pressure through the renin–angiotensin system. ACE inhibitors are used as a first-line medication to be prescribed to treat hypertension, chronic kidney disease, and heart failure, among others. It has been suggested that ACE inhibitors can alleviate the symptoms in mouse models. Despite the benefits of ACE inhibitors, previous studies also have suggested that genetic variants of the ACE gene are risk factors for Alzheimer’s disease (AD) and other neurological diseases, while other variants are associated with reduced risk of AD. In mice, ACE overexpression in the brain reduces symptoms of the AD model systems. Thus, we find two opposing effects of ACE on health. To clarify the effects, we dissect the functions of ACE as follows: (1) angiotensin-converting enzyme that hydrolyzes angiotensin I to make angiotensin II in the renin–angiotensin system; (2) amyloid-degrading enzyme that hydrolyzes beta-amyloid, reducing amyloid toxicity. The efficacy of the ACE inhibitors is well established in humans, while the knowledge specific to AD remains to be open for further research. We provide an overview of ACE and inhibitors that link a wide variety of age-related comorbidities from hypertension to AD to aging. ACE also serves as an example of the middle-life crisis theory that assumes deleterious events during midlife, leading to age-related later events.
Collapse
|
33
|
Kumar M, Bansal N. A Revisit to Etiopathogenesis and Therapeutic Strategies in Alzheimer's Disease. Curr Drug Targets 2021; 23:486-512. [PMID: 34792002 DOI: 10.2174/1389450122666211118125233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 09/05/2021] [Accepted: 09/13/2021] [Indexed: 11/22/2022]
Abstract
Dementia is a cluster of brain abnormalities that trigger progressive memory deficits and other cognitive abilities such as skills, language, or executive function. Alzheimer's disease (AD) is the foremost type of age-associated dementia that involves progressive neurodegeneration accompanied by profound cognitive deficits in advanced stages that severely hamper social or occupational abilities with or without the involvement of any other psychiatric condition. The last two decades witnessed a sharp increase (~123%) in mortality due to AD type dementia, typically owing to a very low disclosure rate (~45%) and hence, the prophylactic, as well as the therapeutic cure of AD, has been a huge challenge. Although understanding of AD pathogenesis has witnessed a remarkable growth (e.g., tauopathy, oxidative stress, lipid transport, glucose uptake, apoptosis, synaptic dysfunction, inflammation, and immune system), still a dearth of an effective therapeutic agent in the management of AD prompts the quest for newer pharmacological targets in the purview of its growing epidemiological status. Most of the current therapeutic strategies focus on modulation of a single target, e.g., inhibition of acetylcholinesterase, glutamate excitotoxicity (memantine), or nootropics (piracetam), even though AD is a multifaceted neurological disorder. There is an impedance urgency to find not only symptomatic but effective disease-modifying therapies. The present review focuses on the risk / protective factors and pathogenic mechanisms involved in AD. In addition to the existing symptomatic therapeutic approach, a diverse array of possible targets linked to pathogenic cascades have been re-investigated to envisage the pharmacotherapeutic strategies in AD.
Collapse
Affiliation(s)
- Manish Kumar
- Chitkara College of Pharmacy, Chitkara University, Punjab. India
| | - Nitin Bansal
- Department of Pharmaceutical Sciences, Chaudhary Bansi Lal University (CBLU), Bhiwani, Haryana 127021. India
| |
Collapse
|
34
|
Chakraborty A, Sami SA, Marma KKS. A comprehensive review on RAGE-facilitated pathological pathways connecting Alzheimer’s disease, diabetes mellitus, and cardiovascular diseases. THE EGYPTIAN JOURNAL OF INTERNAL MEDICINE 2021. [DOI: 10.1186/s43162-021-00081-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Alzheimer’s disease (AD), cardiovascular disease (CVD), and diabetes are some of the most common causes of morbidity and mortality among the aging populations and cause a heavy burden on the worldwide healthcare system. In this review, we briefly highlighted cellular inflammation-based pathways of diabetes mellitus and CVD through receptor for advanced glycation end products AGEs or RAGE leading to Alzheimer’s disease and interrelation between these vascular and metabolic disorders. The articles were retrieved from Google Scholar, Science Direct, and PubMed databases using the following terms: Alzheimer’s; AGEs; RAGE; RAGE in Alzheimer’s; AGEs in Alzheimer’s; RAGE in diabetes; RAGE related pathways of CVD; RAGE in hypertension; RAGE and RAS system; RAGE and oxidative stress.
Main body of the abstract
AD is a neurodegenerative disease characterized by cognitive dysfunction and neuronal cell death. Vascular complications like hypertension, coronary artery disease, and atherosclerosis as well as metabolic syndromes like obesity and diabetes are related to the pathophysiology of AD. RAGE plays significant role in the onset and progression of AD. Amyloid plaques and neurofibrillary tangles (NFT) are two main markers of AD that regulates via RAGE and other RAGE/ligands interactions which also induces oxidative stress and a cascade of other cellular inflammation pathways leading to AD. Though AD and diabetes are two different disorders but may be inter-linked by AGEs and RAGE. In long-term hyperglycemia, upregulated AGEs interacts with RAGE and produces reactive oxygen species which induces further inflammation and vascular complications. Aging, hypercholesterolemia, atherosclerosis, hypertension, obesity, and inflammation are some of the main risk factors for both diabetes and dementia. Chronic hypertension and coronary artery disease disrupt the functions of the blood-brain barrier and are responsible for the accumulation of senile plaques and NFTs.
Short conclusion
RAGE plays a role in the etiology of Aβ and tau hyperphosphorylation, both of which contribute to cognitive impairment. So far, targeting RAGE may provide a potential sight to develop therapies against some metabolic disorders.
Collapse
|
35
|
Ismael S, Mirzahosseini G, Ahmed HA, Yoo A, Kassan M, Malik KU, Ishrat T. Renin-Angiotensin System Alterations in the Human Alzheimer's Disease Brain. J Alzheimers Dis 2021; 84:1473-1484. [PMID: 34690145 DOI: 10.3233/jad-215051] [Citation(s) in RCA: 2] [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 Understanding Alzheimer's disease (AD) in terms of its various pathophysiological pathways is essential to unravel the complex nature of the disease process and identify potential therapeutic targets. The renin-angiotensin system (RAS) has been implicated in several brain diseases, including traumatic brain injury, ischemic stroke, and AD. OBJECTIVE This study was designed to evaluate the protein expression levels of RAS components in postmortem cortical and hippocampal brain samples obtained from AD versus non-AD individuals. METHODS We analyzed RAS components in the cortex and hippocampus of postmortem human brain samples by western blotting and immunohistochemical techniques in comparison with age-matched non-demented controls. RESULTS The expression of AT1R increased in the hippocampus, whereas AT2R expression remained almost unchanged in the cortical and hippocampal regions of AD compared to non-AD brains. The Mas receptor was downregulated in the hippocampus. We also detected slight reductions in ACE-1 protein levels in both the cortex and hippocampus of AD brains, with minor elevations in ACE-2 in the cortex. We did not find remarkable differences in the protein levels of angiotensinogen and Ang II in either the cortex or hippocampus of AD brains, whereas we observed a considerable increase in the expression of brain-derived neurotrophic factor in the hippocampus. CONCLUSION The current findings support the significant contribution of RAS components in AD pathogenesis, further suggesting that strategies focusing on the AT1R and AT2R pathways may lead to novel therapies for the management of AD.
Collapse
Affiliation(s)
- Saifudeen Ismael
- Departments of Anatomy and Neurobiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Golnoush Mirzahosseini
- Departments of Anatomy and Neurobiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA.,Departments of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Heba A Ahmed
- Departments of Anatomy and Neurobiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Arum Yoo
- Departments of Anatomy and Neurobiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Modar Kassan
- Departments of Physiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Kafait U Malik
- Departments of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA.,Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Tauheed Ishrat
- Departments of Anatomy and Neurobiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA.,Departments of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA.,Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
36
|
Sfera A, Osorio C, Maguire G, Rahman L, Afzaal J, Cummings M, Maldonado JC. COVID-19, ferrosenescence and neurodegeneration, a mini-review. Prog Neuropsychopharmacol Biol Psychiatry 2021; 109:110230. [PMID: 33373681 PMCID: PMC7832711 DOI: 10.1016/j.pnpbp.2020.110230] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 12/16/2020] [Accepted: 12/18/2020] [Indexed: 02/06/2023]
Abstract
Exacerbation of cognitive, motor and nonmotor symptoms have been described in critically ill COVID-19 patients, indicating that, like prior pandemics, neurodegenerative sequelae may mark the aftermath of this viral infection. Moreover, SARS-CoV-2, the causative agent of COVID-19 disease, was associated with hyperferritinemia and unfavorable prognosis in older individuals, suggesting virus-induced ferrosenescence. We have previously defined ferrosenescence as an iron-associated disruption of both the human genome and its repair mechanisms, leading to premature cellular senescence and neurodegeneration. As viruses replicate more efficiently in iron-rich senescent cells, they may have developed the ability to induce this phenotype in host tissues, predisposing to both immune dysfunction and neurodegenerative disorders. In this mini-review, we summarize what is known about the SARS-CoV-2-induced cellular senescence and iron dysmetabolism. We also take a closer look at immunotherapy with natural killer cells, angiotensin II receptor blockers ("sartans"), iron chelators and dipeptidyl peptidase 4 inhibitors ("gliptins") as adjunct treatments for both COVID-19 and its neurodegenerative complications.
Collapse
Affiliation(s)
- Adonis Sfera
- Patton State Hospital, California, United States of America.
| | | | - Gerald Maguire
- University of California, Riverside, United States of America
| | - Leah Rahman
- Patton State Hospital, California, United States of America
| | - Jafri Afzaal
- Patton State Hospital, California, United States of America
| | | | | |
Collapse
|
37
|
Rukavina Mikusic NL, Pineda AM, Gironacci MM. Angiotensin-(1-7) and Mas receptor in the brain. EXPLORATION OF MEDICINE 2021. [DOI: 10.37349/emed.2021.00046] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The renin-angiotensin system (RAS) is a key regulator of blood pressure and electrolyte homeostasis. Besides its importance as regulator of the cardiovascular function, the RAS has also been associated to the modulation of higher brain functions, including cognition, memory, depression and anxiety. For many years, angiotensin II (Ang II) has been considered the major bioactive component of the RAS. However, the existence of many other biologically active RAS components has currently been recognized, with similar, opposite, or distinct effects to those exerted by Ang II. Today, it is considered that the RAS is primarily constituted by two opposite arms. The pressor arm is composed by Ang II and the Ang II type 1 (AT1) receptor (AT1R), which mediates the vasoconstrictor, proliferative, hypertensive, oxidative and pro-inflammatory effects of the RAS. The depressor arm is mainly composed by Ang-(1-7), its Mas receptor (MasR) which mediates the depressor, vasodilatory, antiproliferative, antioxidant and anti-inflammatory effects of Ang-(1-7) and the AT2 receptor (AT2R), which opposes to the effects mediated by AT1R activation. Central Ang-(1-7) is implicated in the control of the cardiovascular function, thus participating in the regulation of blood pressure. Ang-(1-7) also exerts neuroprotective actions through MasR activation by opposing to the harmful effects of the Ang II/AT1R axis. This review is focused on the expression and regulation of the Ang-(1-7)/MasR axis in the brain, its main neuroprotective effects and the evidence regarding its involvement in the pathophysiology of several diseases at cardiovascular and neurological level.
Collapse
Affiliation(s)
- Natalia L. Rukavina Mikusic
- Dpto. Química Biológica, IQUIFIB (UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, 1113 Buenos Aires, Argentina
| | - Angélica M. Pineda
- Dpto. Química Biológica, IQUIFIB (UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, 1113 Buenos Aires, Argentina
| | - Mariela M. Gironacci
- Dpto. Química Biológica, IQUIFIB (UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, 1113 Buenos Aires, Argentina
| |
Collapse
|
38
|
Yang W, Luo H, Ma Y, Si S, Zhao H. Effects of Antihypertensive Drugs on Cognitive Function in Elderly Patients with Hypertension: A Review. Aging Dis 2021; 12:841-851. [PMID: 34094646 PMCID: PMC8139194 DOI: 10.14336/ad.2020.1111] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 11/11/2020] [Indexed: 12/13/2022] Open
Abstract
Hypertension is a common comorbidity that contributes to the development of various cardiovascular disorders in elderly patients. Moreover, hypertension has been associated with cognitive decline and dementia. Cognitive impairment leads to increased morbidity and mortality in elderly patients with hypertension. However, previous studies investigating the association between blood pressure (BP), BP variability (BPV), and antihypertensive drug use and the risk of cognitive impairment in elderly patients with hypertension have reported inconsistent findings. Given the global burden of hypertension, the aging population, and the low quality of life associated with cognitive impairment, a more comprehensive understanding of the association between hypertension and cognitive decline is needed. In this review, we summarized the current preclinical evidence and clinical research regarding the association of BP control, BPV, and antihypertensive drug use and cognitive function. We particularly focused on the differences among categories of antihypertensive drugs. We concluded that the correlation of BP and risk of cognitive function is non-linear and dependent on a patient's age. Intensive BP control is generally not recommended, particularly for the oldest-old. Increased BPV and characteristics of orthostatic hypotension in the elderly also increase the risk of cognitive decline. The current evidence does not support one category of antihypertensive drugs as superior to others for preventing dementia in elderly patients with hypertension.
Collapse
Affiliation(s)
- Wei Yang
- Department of Geriatric Medicine, Xuan Wu Hospital, Capital Medical University, Beijing 100053, China
| | - Hongyu Luo
- Department of Geriatric Medicine, Xuan Wu Hospital, Capital Medical University, Beijing 100053, China
| | - Yixin Ma
- Department of Geriatric Medicine, Xuan Wu Hospital, Capital Medical University, Beijing 100053, China
| | - Sicong Si
- Department of Geriatric Medicine, Xuan Wu Hospital, Capital Medical University, Beijing 100053, China
| | - Huan Zhao
- Department of Geriatric Medicine, Xuan Wu Hospital, Capital Medical University, Beijing 100053, China
| |
Collapse
|
39
|
Revi N, Rengan AK. Impact of dietary polyphenols on neuroinflammation-associated disorders. Neurol Sci 2021; 42:3101-3119. [PMID: 33988799 DOI: 10.1007/s10072-021-05303-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 05/05/2021] [Indexed: 12/21/2022]
Abstract
Neurodegenerative disorders like Alzheimer's, Parkinson's, and associated dementia typically originate with altered protein folding and aggregation of their β structures in the neurons. This self-aggregation leads to glial activation in the brain, causing neuroinflammation and leads to neuronal death. According to statistics provided by WHO, there are around 50 million people with dementia worldwide and every year, 10 million more cases are projected to increase. Also, around 5-8 percentage of people who are aged above 60 globally has dementia or associated disorders. Over 82 million in 2030 and 152 in 2050 are expected to have dementia. Most of these patients fall into low-middle-income countries which makes it even more essential to find an affordable and effective treatment method. Polyphenols of different origin are studied for their potential role as anti-neuro-inflammatory molecules. This review would summarize recent advances in three widely researched dietary polyphenols projected as potential therapeutic agents for disorders like Alzheimer's, Parkinson's, etc. They are Resveratrol, Catechins, and Tannins. The review would discuss the recent advances and challenges in using these polyphenols using specific examples as potential therapeutic agents against neuroinflammation associated disorders. An abstract of neuroinflammation-associated events and the effects by selected polyphenols.
Collapse
Affiliation(s)
- Neeraja Revi
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, India
| | - Aravind Kumar Rengan
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, India.
| |
Collapse
|
40
|
Gamba P, Giannelli S, Staurenghi E, Testa G, Sottero B, Biasi F, Poli G, Leonarduzzi G. The Controversial Role of 24-S-Hydroxycholesterol in Alzheimer's Disease. Antioxidants (Basel) 2021; 10:antiox10050740. [PMID: 34067119 PMCID: PMC8151638 DOI: 10.3390/antiox10050740] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 04/29/2021] [Accepted: 05/03/2021] [Indexed: 01/19/2023] Open
Abstract
The development of Alzheimer’s disease (AD) is influenced by several events, among which the dysregulation of cholesterol metabolism in the brain plays a major role. Maintenance of brain cholesterol homeostasis is essential for neuronal functioning and brain development. To maintain the steady-state level, excess brain cholesterol is converted into the more hydrophilic metabolite 24-S-hydroxycholesterol (24-OHC), also called cerebrosterol, by the neuron-specific enzyme CYP46A1. A growing bulk of evidence suggests that cholesterol oxidation products, named oxysterols, are the link connecting altered cholesterol metabolism to AD. It has been shown that the levels of some oxysterols, including 27-hydroxycholesterol, 7β-hydroxycholesterol and 7-ketocholesterol, significantly increase in AD brains contributing to disease progression. In contrast, 24-OHC levels decrease, likely due to neuronal loss. Among the different brain oxysterols, 24-OHC is certainly the one whose role is most controversial. It is the dominant oxysterol in the brain and evidence shows that it represents a signaling molecule of great importance for brain function. However, numerous studies highlighted the potential role of 24-OHC in favoring AD development, since it promotes neuroinflammation, amyloid β (Aβ) peptide production, oxidative stress and cell death. In parallel, 24-OHC has been shown to exert several beneficial effects against AD progression, such as preventing tau hyperphosphorylation and Aβ production. In this review we focus on the current knowledge of the controversial role of 24-OHC in AD pathogenesis, reporting a detailed overview of the findings about its levels in different AD biological samples and its noxious or neuroprotective effects in the brain. Given the relevant role of 24-OHC in AD pathophysiology, its targeting could be useful for disease prevention or slowing down its progression.
Collapse
|
41
|
Basmadjian OM, Occhieppo VB, Marchese NA, Silvero C MJ, Becerra MC, Baiardi G, Bregonzio C. Amphetamine Induces Oxidative Stress, Glial Activation and Transient Angiogenesis in Prefrontal Cortex via AT 1-R. Front Pharmacol 2021; 12:647747. [PMID: 34012397 PMCID: PMC8126693 DOI: 10.3389/fphar.2021.647747] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 03/30/2021] [Indexed: 01/21/2023] Open
Abstract
Background: Amphetamine (AMPH) alters neurons, glia and microvessels, which affects neurovascular unit coupling, leading to disruption in brain functions such as attention and working memory. Oxidative stress plays a crucial role in these alterations. The angiotensin type I receptors (AT1-R) mediate deleterious effects, such as oxidative/inflammatory responses, endothelial dysfunction, neuronal oxidative damage, alterations that overlap with those observed from AMPH exposure. Aims: The aim of this study was to evaluate the AT1-R role in AMPH-induced oxidative stress and glial and vascular alterations in the prefrontal cortex (PFC). Furthermore, we aimed to evaluate the involvement of AT1-R in the AMPH-induced short-term memory and working memory deficit. Methods: Male Wistar rats were repeatedly administered with the AT1-R blocker candesartan (CAND) and AMPH. Acute oxidative stress in the PFC was evaluated immediately after the last AMPH administration by determining lipid and protein peroxidation. After 21 off-drug days, long-lasting alterations in the glia, microvessel architecture and to cognitive tasks were evaluated by GFAP, CD11b and von Willebrand immunostaining and by short-term and working memory assessment. Results: AMPH induced acute oxidative stress, long-lasting glial reactivity in the PFC and a working memory deficit that were prevented by AT1-R blockade pretreatment. Moreover, AMPH induces transient angiogenesis in PFC via AT1-R. AMPH did not affect short-term memory. Conclusion: Our results support the protective role of AT1-R blockade in AMPH-induced oxidative stress, transient angiogenesis and long-lasting glial activation, preserving working memory performance.
Collapse
Affiliation(s)
- Osvaldo M Basmadjian
- Departamento de Farmacología, Facultad de Ciencias Químicas, Instituto de Farmacología Experimental Córdoba (IFEC-CONICET), Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Victoria B Occhieppo
- Departamento de Farmacología, Facultad de Ciencias Químicas, Instituto de Farmacología Experimental Córdoba (IFEC-CONICET), Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Natalia A Marchese
- Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC), CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina.,Departamento de Química Biológica "Ranwel Caputto", Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - M Jazmin Silvero C
- Instituto Multidisciplinario de Biología Vegetal (IMBIV-CONICET) Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - María Cecilia Becerra
- Instituto Multidisciplinario de Biología Vegetal (IMBIV-CONICET) Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Gustavo Baiardi
- Laboratorio de Neurofarmacología, (IIBYT-CONICET), Universidad Nacional de Córdoba, Córdoba, Argentina.,Facultad de Ciencias Químicas, Universidad Católica de Córdoba, Córdoba, Argentina
| | - Claudia Bregonzio
- Departamento de Farmacología, Facultad de Ciencias Químicas, Instituto de Farmacología Experimental Córdoba (IFEC-CONICET), Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
42
|
Ranjit A, Khajehpour S, Aghazadeh-Habashi A. Update on Angiotensin II Subtype 2 Receptor: Focus on Peptide and Nonpeptide Agonists. Mol Pharmacol 2021; 99:469-487. [PMID: 33795351 DOI: 10.1124/molpharm.121.000236] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 03/12/2021] [Indexed: 11/22/2022] Open
Abstract
Angiotensin II (Ang II) is the most dominant effector component of the renin-angiotensin system (RAS) that generally acts through binding to two main classes of G protein-coupled receptors, namely Ang II subtype 1 receptor (AT1R) and angiotensin II subtype 2 receptor (AT2R). Despite some controversial reports, the activation of AT2R generally antagonizes the effects of Ang II binding on AT1R. Studying AT2R signaling, function, and its specific ligands in cell culture or animal studies has confirmed its beneficial effects throughout the body. These characteristics classify AT2R as part of the protective arm of the RAS that, along with functions of Ang (1-7) through Mas receptor signaling, modulates the harmful effects of Ang II on AT1R in the activated classic arm of the RAS. Although Ang II is the primary ligand for AT2R, we have summarized other natural or synthetic peptide and nonpeptide agonists with critical evaluation of their structure, mechanism of action, and biologic activity. SIGNIFICANCE STATEMENT: AT2R is one of the main components of the RAS and has a significant prospective for mediating the beneficial action of the RAS through its protective arm on the body's homeostasis. Targeting AT2R offers substantial clinical application possibilities for modulating various pathological conditions. This review provided concise information regarding the AT2R peptide and nonpeptide agonists and their potential clinical applications for various diseases.
Collapse
Affiliation(s)
- Arina Ranjit
- College of Pharmacy, Idaho State University, Pocatello, Idaho, USA
| | - Sana Khajehpour
- College of Pharmacy, Idaho State University, Pocatello, Idaho, USA
| | | |
Collapse
|
43
|
Chin LY, Tan JYP, Choudhury H, Pandey M, Sisinthy SP, Gorain B. Development and optimization of chitosan coated nanoemulgel of telmisartan for intranasal delivery: A comparative study. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102341] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
44
|
Dhangadamajhi G, Singh S. Malaria link of hypertension: a hidden syndicate of angiotensin II, bradykinin and sphingosine 1-phosphate. Hum Cell 2021; 34:734-744. [PMID: 33683655 DOI: 10.1007/s13577-021-00513-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 02/22/2021] [Indexed: 01/22/2023]
Abstract
In malaria-endemic countries, the burden of hypertension is on the rise. Although malaria and hypertension seem to have no direct link, several studies in recent years support their possible link. Three bioactive molecules such as angiotensin II (Ang II), bradykinin (BK) and sphingosine 1-phosphate (S1P) are crucial in regulating blood pressure. While the increased level of Ang II and S1P are responsible for inducing hypertension, BK is arthero-protective and anti-hypertensive. Therefore, in the present review, based on available literatures we highlight the present knowledge on the production and bioavailability of these molecules, the mechanism of their regulation of hypertension, and patho-physiological role in malaria. Further, a possible link between malaria and hypertension is hypothesized through various arguments based on experimental evidence. Understanding of their mechanisms of blood pressure regulation during malaria infection may open up avenues for drug therapeutics and management of malaria in co-morbidity with hypertension.
Collapse
Affiliation(s)
- Gunanidhi Dhangadamajhi
- Department of Biotechnology, Maharaja Sriramchandra Bhanjadeo University, Takatpur, Baripada, Odisha, 75003, India.
| | - Shailja Singh
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| |
Collapse
|
45
|
Ziaja M, Urbanek KA, Kowalska K, Piastowska-Ciesielska AW. Angiotensin II and Angiotensin Receptors 1 and 2-Multifunctional System in Cells Biology, What Do We Know? Cells 2021; 10:cells10020381. [PMID: 33673178 PMCID: PMC7917773 DOI: 10.3390/cells10020381] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/05/2021] [Accepted: 02/09/2021] [Indexed: 12/13/2022] Open
Abstract
For years, the renin-angiotensin system (RAS) has been perceived as a system whose role is to primarily modulate the functioning of the cardiovascular system. Years of research into the role of RAS have provided the necessary data to confirm that the role of RAS is very complex and not limited to the cardiovascular system. The presence of individual elements of the renin-angiotensin (RA) system allows to control many processes, ranging from the memorization to pro-cancer processes. Maintaining the proportions between the individual axes of the RA system allows for achieving a balance, often called homeostasis. Thus, any disturbance in the expression or activity of individual RAS elements leads to pathophysiological processes.
Collapse
|
46
|
Ouk M, Wu CY, Rabin JS, Jackson A, Edwards JD, Ramirez J, Masellis M, Swartz RH, Herrmann N, Lanctôt KL, Black SE, Swardfager W. The use of angiotensin-converting enzyme inhibitors vs. angiotensin receptor blockers and cognitive decline in Alzheimer's disease: the importance of blood-brain barrier penetration and APOE ε4 carrier status. ALZHEIMERS RESEARCH & THERAPY 2021; 13:43. [PMID: 33573702 PMCID: PMC7876820 DOI: 10.1186/s13195-021-00778-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 01/25/2021] [Indexed: 12/15/2022]
Abstract
Background The antihypertensive angiotensin receptor blockers (ARBs) and angiotensin-converting enzyme inhibitors (ACE-Is) have similar indications and mechanisms of action, but prior work suggests divergence in their effects on cognition. Methods Participants in the National Alzheimer’s Coordinating Center database with a clinical diagnosis of dementia due to Alzheimer’s disease (AD) using an ACE-I or an ARB at any visit were selected. The primary outcome was delayed recall memory on the Wechsler Memory Scale Revised – Logical Memory IIA. Other cognitive domains were explored, including attention and psychomotor processing speed (Trail Making Test [TMT]-A and Digit Symbol Substitution Test [DSST]), executive function (TMT-B), and language and semantic verbal fluency (Animal Naming, Vegetable Naming, and Boston Naming Tests). Random slopes mixed-effects models with inverse probability of treatment weighting were used, yielding rate ratios (RR) or regression coefficients (B), as appropriate to the distribution of the data. Apolipoprotein (APOE) ε4 status and blood-brain barrier (BBB) penetrance were investigated as effect modifiers. Results Among 1689 participants with AD, ARB use (n = 578) was associated with 9.4% slower decline in delayed recall performance over a mean follow-up of 2.28 years compared with ACE-I use (n = 1111) [RR = 1.094, p = 0.0327]; specifically, users of BBB-crossing ARBs (RR = 1.25, p = 0.002), BBB-crossing ACE-Is (RR = 1.16, p = 0.010), and non-BBB-crossing ARBs (RR = 1.20, p = 0.005) had better delayed recall performance over time compared with non-BBB-crossing ACE-I users. An interaction with APOE ε4 status (drug × APOE × time RR = 1.196, p = 0.033) emerged; ARBs were associated with better delayed recall scores over time than ACE-Is in non-carriers (RR = 1.200, p = 0.003), but not in carriers (RR = 1.003, p = 0.957). ARB use was also associated with better performance over time on the TMT-A (B = 2.023 s, p = 0.0004) and the DSST (B = 0.573 symbols, p = 0.0485), and these differences were significant among APOE ε4 non-carriers (B = 4.066 s, p = 0.0004; and B = 0.982 symbols, p = 0.0230; respectively). Some differences were seen also in language and verbal fluency among APOE ε4 non-carriers. Conclusions Among APOE ε4 non-carriers with AD, ARB use was associated with greater preservation of memory and attention/psychomotor processing speed, particularly compared to ACE-Is that do not cross the blood-brain-barrier. Supplementary Information The online version contains supplementary material available at 10.1186/s13195-021-00778-8.
Collapse
Affiliation(s)
- Michael Ouk
- Department of Pharmacology & Toxicology Room 4207, University of Toronto, Medical Sciences Building 1 King's College Circle, Toronto, ON, M5S 1A8, Canada.,Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Che-Yuan Wu
- Department of Pharmacology & Toxicology Room 4207, University of Toronto, Medical Sciences Building 1 King's College Circle, Toronto, ON, M5S 1A8, Canada.,Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Jennifer S Rabin
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada.,Harquail Centre for Neuromodulation, Sunnybrook Research Institute, Toronto, M4N 3M5, Canada.,Department of Medicine (Neurology), Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada.,Rehabilitation Sciences Institute, University of Toronto, Toronto, ON, Canada
| | - Aaron Jackson
- Department of Pharmacology & Toxicology Room 4207, University of Toronto, Medical Sciences Building 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
| | - Jodi D Edwards
- University of Ottawa Heart Institute, University of Ottawa, Ottawa, ON, Canada.,School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON, Canada.,ICES, Ottawa, ON, Canada
| | - Joel Ramirez
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada.,Canadian Partnership for Stroke Recovery, Toronto, ON, Canada
| | - Mario Masellis
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada.,Department of Medicine (Neurology), Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Richard H Swartz
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada.,Department of Medicine (Neurology), Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada.,Canadian Partnership for Stroke Recovery, Toronto, ON, Canada
| | - Nathan Herrmann
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada.,Department of Psychiatry, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Krista L Lanctôt
- Department of Pharmacology & Toxicology Room 4207, University of Toronto, Medical Sciences Building 1 King's College Circle, Toronto, ON, M5S 1A8, Canada.,Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada.,Department of Psychiatry, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada.,KITE UHN Toronto Rehabilitation Institute, Toronto, ON, Canada
| | - Sandra E Black
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada.,School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON, Canada.,Canadian Partnership for Stroke Recovery, Toronto, ON, Canada.,KITE UHN Toronto Rehabilitation Institute, Toronto, ON, Canada
| | - Walter Swardfager
- Department of Pharmacology & Toxicology Room 4207, University of Toronto, Medical Sciences Building 1 King's College Circle, Toronto, ON, M5S 1A8, Canada. .,Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada. .,Canadian Partnership for Stroke Recovery, Toronto, ON, Canada. .,KITE UHN Toronto Rehabilitation Institute, Toronto, ON, Canada.
| |
Collapse
|
47
|
Ribeiro VT, de Souza LC, Simões E Silva AC. Renin-Angiotensin System and Alzheimer's Disease Pathophysiology: From the Potential Interactions to Therapeutic Perspectives. Protein Pept Lett 2020; 27:484-511. [PMID: 31886744 DOI: 10.2174/0929866527666191230103739] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 08/27/2019] [Accepted: 11/16/2019] [Indexed: 12/21/2022]
Abstract
New roles of the Renin-Angiotensin System (RAS), apart from fluid homeostasis and Blood Pressure (BP) regulation, are being progressively unveiled, since the discoveries of RAS alternative axes and local RAS in different tissues, including the brain. Brain RAS is reported to interact with pathophysiological mechanisms of many neurological and psychiatric diseases, including Alzheimer's Disease (AD). Even though AD is the most common cause of dementia worldwide, its pathophysiology is far from elucidated. Currently, no treatment can halt the disease course. Successive failures of amyloid-targeting drugs have challenged the amyloid hypothesis and increased the interest in the inflammatory and vascular aspects of AD. RAS compounds, both centrally and peripherally, potentially interact with neuroinflammation and cerebrovascular regulation. This narrative review discusses the AD pathophysiology and its possible interaction with RAS, looking forward to potential therapeutic approaches. RAS molecules affect BP, cerebral blood flow, neuroinflammation, and oxidative stress. Angiotensin (Ang) II, via angiotensin type 1 receptors may promote brain tissue damage, while Ang-(1-7) seems to elicit neuroprotection. Several studies dosed RAS molecules in AD patients' biological material, with heterogeneous results. The link between AD and clinical conditions related to classical RAS axis overactivation (hypertension, heart failure, and chronic kidney disease) supports the hypothesized role of this system in AD. Additionally, RAStargeting drugs as Angiotensin Converting Enzyme inhibitors (ACEis) and Angiotensin Receptor Blockers (ARBs) seem to exert beneficial effects on AD. Results of randomized controlled trials testing ACEi or ARBs in AD are awaited to elucidate whether AD-RAS interaction has implications on AD therapeutics.
Collapse
Affiliation(s)
- Victor Teatini Ribeiro
- Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Leonardo Cruz de Souza
- Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil.,Department of Internal Medicine, Service of Neurology, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Ana Cristina Simões E Silva
- Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| |
Collapse
|
48
|
Armstrong TD, Suwannasual U, Kennedy CL, Thasma A, Schneider LJ, Phillippi D, Lund AK. Exposure to Traffic-Generated Pollutants Exacerbates the Expression of Factors Associated with the Pathophysiology of Alzheimer’s Disease in Aged C57BL/6 Wild-Type Mice. J Alzheimers Dis 2020; 78:1453-1471. [DOI: 10.3233/jad-200929] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Background: Multiple studies report a strong correlation between traffic-generated air pollution-exposure and detrimental outcomes in the central nervous system (CNS), including Alzheimer’s disease (AD). Incidence of AD is rapidly increasing and, worldwide, many live in regions where pollutants exceed regulatory standards. Thus, it is imperative to identify environmental pollutants that contribute to AD, and the mechanisms involved. Objective: We investigated the effects of mixed gasoline and diesel engine emissions (MVE) on the expression of factors involved in progression of AD in the hippocampus and cerebrum in a young versus aged mouse model. Methods: Young (2 months old) and aged (18 months old) male C57BL/6 mice were exposed to either MVE (300μg/m3 PM) or filtered air (FA) for 6 h/d, 7 d/wk, for 50 d. Immunofluorescence and RT-qPCR were used to quantify oxidative stress (8-OHdG) and expression of amyloid-β protein precursor (AβPP), β secretase (BACE1), amyloid-β (Aβ), aryl hydrocarbon receptor (AhR), cytochrome P450 (CYP) 1B1, angiotensin-converting enzyme (ACE1), and angiotensin II type 1 (AT1) receptor in the cerebrum and hippocampus, in addition to cerebral microvascular tight junction (TJ) protein expression. Results: We observed age-related increases in oxidative stress, AhR, CYP1B1, Aβ, BACE1, and AT1 receptor in the CA1 region of the hippocampus, and elevation of cerebral AβPP, AhR, and CYP1B1 mRNA, associated with decreased cerebral microvascular TJ protein claudin-5. MVE-exposure resulted in further promotion of oxidative stress, and significant increases in AhR, CYP1B1, BACE1, ACE1, and Aβ, compared to the young and aged FA-exposed mice. Conclusion: Such findings suggest that MVE-exposure exacerbates the expression of factors in the CNS associated with AD pathogenesis in aged populations.
Collapse
Affiliation(s)
- Tyler D. Armstrong
- Advanced Environmental Research Institute, Department of Biological Sciences, University of North Texas, Denton, TX, USA
| | - Usa Suwannasual
- Advanced Environmental Research Institute, Department of Biological Sciences, University of North Texas, Denton, TX, USA
| | - Conner L. Kennedy
- Advanced Environmental Research Institute, Department of Biological Sciences, University of North Texas, Denton, TX, USA
| | - Akshaykumar Thasma
- Advanced Environmental Research Institute, Department of Biological Sciences, University of North Texas, Denton, TX, USA
| | - Leah J. Schneider
- Advanced Environmental Research Institute, Department of Biological Sciences, University of North Texas, Denton, TX, USA
| | - Danielle Phillippi
- Advanced Environmental Research Institute, Department of Biological Sciences, University of North Texas, Denton, TX, USA
| | - Amie K. Lund
- Advanced Environmental Research Institute, Department of Biological Sciences, University of North Texas, Denton, TX, USA
| |
Collapse
|
49
|
Salminen A. Hypoperfusion is a potential inducer of immunosuppressive network in Alzheimer's disease. Neurochem Int 2020; 142:104919. [PMID: 33242538 DOI: 10.1016/j.neuint.2020.104919] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 10/12/2020] [Accepted: 11/19/2020] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease which causes a non-reversible cognitive impairment and dementia. The primary cause of late-onset AD remains unknown although its pathology was discovered over a century ago. Recently, the vascular hypothesis of AD has received backing from evidence emerging from neuroimaging studies which have revealed the presence of a significant hypoperfusion in the brain regions vulnerable to AD pathology. In fact, hypoxia can explain many of the pathological changes evident in AD pathology, e.g. the deposition of β-amyloid plaques and chronic low-grade inflammation. Hypoxia-inducible factor-1α (HIF-1α) stimulates inflammatory responses and modulates both innate and adaptive immunity. It is known that hypoxia-induced inflammation evokes compensatory anti-inflammatory response involving tissue-resident microglia/macrophages and infiltrated immune cells. Hypoxia/HIF-1α induce immunosuppression by (i) increasing the expression of immunosuppressive genes, (ii) stimulating adenosinergic signaling, (iii) enhancing aerobic glycolysis, i.e. lactate production, and (iv) augmenting the secretion of immunosuppressive exosomes. Interestingly, it seems that these common mechanisms are also involved in the pathogenesis of AD. In AD pathology, an enhanced immunosuppression appears, e.g. as a shift in microglia/macrophage phenotypes towards the anti-inflammatory M2 phenotype and an increase in the numbers of regulatory T cells (Treg). The augmented anti-inflammatory capacity promotes the resolution of acute inflammation but persistent inflammation has crucial effects not only on immune cells but also harmful responses to the homeostasis of AD brain. I will examine in detail the mechanisms of the hypoperfusion/hypoxia-induced immunosuppressive state in general and especially, in its association with AD pathogenesis. These immunological observations support the vascular hypothesis of AD pathology.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland.
| |
Collapse
|
50
|
Li X, Xuan W, Chen D, Gao H, Wang G, Guo Q, Wang Y, Song H, Cai B. Research Progress of Alzheimer's Disease Therapeutic Drugs: Based on Renin-Angiotensin System Axis. J Alzheimers Dis 2020; 78:1315-1338. [PMID: 33164932 DOI: 10.3233/jad-200770] [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: 11/15/2022]
Abstract
It is widely recognized that Alzheimer's disease (AD) has a complicate link to renin-angiotensin system (RAS). It is known that cerebrovascular disease has some connections with AD, but most of the studies are still conducted in parallel or independently. Although previous research came up with large number of hypotheses about the pathogenesis of AD, it does not include the mechanism of RAS-related regulation of AD. It has been found that many components of RAS have been changed in AD. For example, the multifunctional and high-efficiency vasoconstrictor Ang II and Ang III with similar effects are changed under the action of other RAS signal peptides; these signal peptides are believed to help improve nerve injury and cognitive function. These changes may lead to neuropathological changes of AD, and progressive defects of cognitive function, which are association with some hypotheses of AD. The role of RAS in AD gradually attracts our attention, and RAS deserved to be considered carefully in the pathogenesis of AD. This review discusses the mechanisms of RAS participating in the three current hypotheses of AD: neuroinflammation, oxidative stress and amyloid-β protein (Aβ) hypothesis, as well as the drugs that regulate RAS systems already in clinical or in clinical trials. It further demonstrates the importance of RAS in the pathogenesis of AD, not only because of its multiple aspects of participation, which may be accidental, but also because of the availability of RAS drugs, which can be reused as therapies of AD.
Collapse
Affiliation(s)
- Xinquan Li
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Weiting Xuan
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Dabao Chen
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Huawu Gao
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Guangyun Wang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Qiaoru Guo
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the Fifth Affiliated Hospital and School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yan Wang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Hang Song
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Biao Cai
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| |
Collapse
|