1
|
Lin CY, Chen PH, Tsai CL, Hsieh YW, Hu KC, Tsai FJ, Cho DY, Liao HY. Antihypertensive medication and dementia risk in patients with hypertension: A nationwide population-based study. J Clin Neurosci 2024; 125:83-94. [PMID: 38759352 DOI: 10.1016/j.jocn.2024.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 04/27/2024] [Accepted: 05/07/2024] [Indexed: 05/19/2024]
Abstract
The claim between hypertension and dementia needs more evidence due to limited data. We aim to examine the risk of dementia in patients with hypertension, and determine whether the use of antihypertensive medications (AHMs) could decrease the incidence of dementia diagnosed following the onset of hypertension. We employed the Taiwan National Health Insurance Research Database from 2000 to 2016 and performed a retrospective cohort study. We also carried out a case-control study to see if AHMs could reduce the incidence of newly diagnosed dementia in hypertensive patients. In the retrospective cohort study, we selected 587,762 participants with age and gender matched in experimental and control groups. The hypertension group had significantly higher adjusted hazard ratios (aHRs) of getting newly diagnosed dementia, including all-cause dementia, Alzheimer's disease, and vascular dementia (aHR, 2.86; 95 % confidence interval (CI), 2.74-2.99) than the control group. Three kinds of specific AHMs, namely, angiotensin II receptor blockers (aHR, 0.55; 95 % CI, 0.53-0.57), calcium channel blockers (aHR, 0.76; 95 % CI, 0.73-0.80), and diuretics (aHR,0.93; 95 % CI, 0.89-0.97) could significantly decrease the incidence of getting newly diagnosed dementia. Also, the application of traditional Chinese medicine (TCM) significantly associates with the lower aHRs of newly diagnosed dementia in hypertensive patients compared to patients without TCM (aHR, 0.90; 95 % CI, 0.81-1.00). Hypertension may be a significant risk factor for dementia. Both AHMs and TCM significantly associate with the lower incidence of newly diagnosed dementia in hypertensive patients.
Collapse
Affiliation(s)
- Chih-Ying Lin
- Department of Chinese Traumatology Medicine, China Medical University Hospital, Taichung 40402, Taiwan
| | - Pei-Hsien Chen
- Department of Chinese Medicine, China Medical University Hospital, Taichung 40402, Taiwan
| | - Chiu-Lin Tsai
- Department of Chinese Medicine Pharmacy, China Medical University Hospital, Taichung 40402, Taiwan
| | - Yow-Wen Hsieh
- Graduate Institute of Pharmacy, China Medical University, Taichung 40402, Taiwan; Department of Pharmacy, China Medical University Hospital, Taichung 40402, Taiwan
| | - Kai-Chieh Hu
- Management Office for Health Data (Dry Lab), Clinical Trial Center, China Medical University Hospital, Taichung 40402, Taiwan
| | - Fuu-Jen Tsai
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung 40402, Taiwan; Department of Medical Research, China Medical University Hospital, Taichung 40402, Taiwan; Division of Medical Genetics, China Medical University Children's Hospital, Taichung 40447, Taiwan; Department of Biotechnology and Bioinformatics, Asia University, Taichung, Taiwan
| | - Der-Yang Cho
- Translational Cell Therapy Center, Department of Medical Research, China Medical University Hospital, Taichung 40447, Taiwan; Department of Neurosurgery, China Medical University Hospital, Taichung 40447, Taiwan; Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40447, Taiwan
| | - Hsien-Yin Liao
- School of Post-Baccalaureate Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung 40402, Taiwan; Department of Acupuncture, China Medical University Hospital, Taichung 40402, Taiwan.
| |
Collapse
|
2
|
Wells RG, Azzam AF, Hiller AL, Sardinia MF. Effects of an Angiotensin IV Analog on 3-Nitropropionic Acid-Induced Huntington's Disease-Like Symptoms in Rats. J Huntingtons Dis 2024; 13:55-66. [PMID: 38489193 DOI: 10.3233/jhd-231507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2024]
Abstract
Background Huntington's disease (HD) is a neurodegenerative disorder characterized by motor, cognitive, and psychiatric dysfunction caused by a mutant huntingtin protein. Compromised metabolic activity resulting from systemic administration of the mitochondrial toxin, 3-nitropropionic acid (3-NP), is known to mimic the pathology of HD and induce HD-like symptoms in rats. N-hexanoic-Tyr-Ile-(6)-amino hexanoic amide (PNB-0408), also known as Dihexa, has been shown to have neuroprotective and procognitive properties in animal models of Alzheimer's and Parkinson's diseases. Given the mechanism of action and success in other neurodegenerative diseases, we felt it an appropriate compound to investigate further for HD. Objective The present study was designed to test if PNB-0408, an angiotensin IV analog, could attenuate 3-NP-induced HD-like symptoms in rats and serve as a potential therapeutic agent. Methods Forty male Wistar rats were randomized into three groups consisting of a "vehicle" group, a "3-NP" group, and a "3-NP + PNB-0408" group. PNB-0408 was administered along with chronic exposure to 3-NP. Animal body weight, motor function, and cognitive abilities were measured for five weeks, before euthanasia and histopathological analysis. Results Exposure to 3-NP decreased the amount of weight rats gained, impaired spatial learning and memory consolidation, and led to marked motor dysfunction. From our observations and analysis, PNB-0408 did not protect rats from the deficits induced by 3-NP neurotoxicity. Conclusions Our findings suggest that PNB-0408 may not be an efficacious treatment strategy for preventing 3-NP-induced HD-like symptoms in a preclinical model. These data highlight the need for further research of this compound in alternate models and/or alternative approaches to managing this disorder.
Collapse
Affiliation(s)
- Russell G Wells
- Department of Biology, Whitworth University, Spokane, WA, USA
- School of Medicine, Oregon Health and Science University, Portland, OR, USA
| | - Azzam F Azzam
- Department of Biology, Whitworth University, Spokane, WA, USA
- School of Medicine, University of Washington, Seattle, WA, USA
| | - Amie L Hiller
- School of Medicine, Oregon Health and Science University, Portland, OR, USA
| | | |
Collapse
|
3
|
Cosarderelioglu C, Nidadavolu LS, George CJ, Marx-Rattner R, Powell L, Xue QL, Tian J, Oh ES, Ferrucci L, Dincer P, Bennett DA, Walston JD, Abadir PM. Angiotensin receptor blocker use is associated with upregulation of the memory-protective angiotensin type 4 receptor (AT 4R) in the postmortem brains of individuals without cognitive impairment. GeroScience 2023; 45:371-384. [PMID: 35969296 PMCID: PMC9886717 DOI: 10.1007/s11357-022-00639-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 08/04/2022] [Indexed: 02/03/2023] Open
Abstract
The reported primary dementia-protective benefits of angiotensin II type 1 receptor (AT1R) blockers (ARB) are believed, at least in part, to arise from systemic effects on blood pressure. However, there is a specific and independently regulated brain renin-angiotensin system (RAS). Brain RAS acts mainly through three receptor subtypes; AT1R, AT2R, and AT4R. The AT1R promotes inflammation and mitochondrial reactive oxygen species generation. AT2R increases nitric oxide. AT4R is essential for dopamine and acetylcholine release. It is unknown whether ARB use is associated with changes in the brain RAS. Here, we compared the impact of treatment with ARB on not cognitively impaired individuals and individuals with Alzheimer's dementia using postmortem frontal-cortex samples of age- and sex-matched participants (70-90 years old, n = 30 in each group). We show that ARB use is associated with higher brain AT4R, lower oxidative stress, and amyloid-β burden in NCI participants. In AD, ARB use was associated with lower brain AT1R but had no impact on inflammation, oxidative stress, or amyloid-β burden. Our results may suggest a potential role for AT4R in the salutary effects for ARB on the brains of not cognitively impaired older adults.
Collapse
Affiliation(s)
- Caglar Cosarderelioglu
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, MD, 21224, 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, MD, 21224, USA
| | - Claudene J George
- Department of Medicine, Division of Geriatrics, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
| | - Ruth Marx-Rattner
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA
| | - Laura Powell
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA
| | - Qian-Li Xue
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA
- Johns Hopkins University Center On Aging and Health, Baltimore, MD, USA
| | - Jing Tian
- Department of Biostatistics, Bloomberg School of Public Health, Baltimore, MD, USA
| | - Esther S Oh
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA
| | - Luigi Ferrucci
- National Institute On Aging, National Institutes of Health, Baltimore, MD, 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, IL, USA
| | - Jeremy D Walston
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA
| | - Peter M Abadir
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA.
| |
Collapse
|
4
|
Bild W, Vasincu A, Rusu RN, Ababei DC, Stana AB, Stanciu GD, Savu B, Bild V. Impact of the Renin-Angiotensin System on the Pathogeny and Pharmacotherapeutics of Neurodegenerative Diseases. Biomolecules 2022; 12:1429. [PMID: 36291638 PMCID: PMC9599929 DOI: 10.3390/biom12101429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 09/28/2022] [Accepted: 10/03/2022] [Indexed: 11/17/2022] Open
Abstract
Brain neurodegenerative diseases (BND) are debilitating conditions that are especially characteristic of a certain period of life and considered major threats to human health. Current treatments are limited, meaning that there is a challenge in developing new options that can efficiently tackle the different components and pathophysiological processes of these conditions. The renin-angiotensin-aldosterone system (RAS) is an endocrine axis with important peripheral physiological functions such as blood pressure and cardiovascular homeostasis, as well as water and sodium balance and systemic vascular resistance-functions which are well-documented. However, recent work has highlighted the paracrine and autocrine functions of RAS in different tissues, including the central nervous system (CNS). It is known that RAS hyperactivation has pro-inflammatory and pro-oxidant effects, thus suggesting that its pharmacological modulation could be used in the management of these conditions. The present paper underlines the involvement of RAS and its components in the pathophysiology of BNDs such as Parkinson's disease (PD), Alzheimer's disease (AD), multiple sclerosis (MS), Huntington's disease (HD), motor neuron disease (MND), and prion disease (PRD), as well as the identification of drugs and pharmacologically active substances that act upon RAS, which could alleviate their symptomatology or evolution, and thus, contribute to novel therapeutic approaches.
Collapse
Affiliation(s)
- Walther Bild
- Department of Physiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Center of Biomedical Research of the Romanian Academy, 700506 Iasi, Romania
| | - Alexandru Vasincu
- Department of Pharmacodynamics and Clinical Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Răzvan-Nicolae Rusu
- Department of Pharmacodynamics and Clinical Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Daniela-Carmen Ababei
- Department of Pharmacodynamics and Clinical Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Aurelian Bogdan Stana
- Department of Physiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Gabriela Dumitrița Stanciu
- Center for Advanced Research and Development in Experimental Medicine (CEMEX), “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Bogdan Savu
- Department of Pediatric Surgery, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Veronica Bild
- Center of Biomedical Research of the Romanian Academy, 700506 Iasi, Romania
- Department of Pharmacodynamics and Clinical Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Center for Advanced Research and Development in Experimental Medicine (CEMEX), “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| |
Collapse
|
5
|
Méndez-García LA, Escobedo G, Minguer-Uribe AG, Viurcos-Sanabria R, Aguayo-Guerrero JA, Carrillo-Ruiz JD, Solleiro-Villavicencio H. Role of the renin-angiotensin system in the development of COVID-19-associated neurological manifestations. Front Cell Neurosci 2022; 16:977039. [PMID: 36187294 PMCID: PMC9523599 DOI: 10.3389/fncel.2022.977039] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 08/26/2022] [Indexed: 01/18/2023] Open
Abstract
SARS-CoV-2 causes COVID-19, which has claimed millions of lives. This virus can infect various cells and tissues, including the brain, for which numerous neurological symptoms have been reported, ranging from mild and non-life-threatening (e.g., headaches, anosmia, dysgeusia, and disorientation) to severe and life-threatening symptoms (e.g., meningitis, ischemic stroke, and cerebral thrombosis). The cellular receptor for SARS-CoV-2 is angiotensin-converting enzyme 2 (ACE2), an enzyme that belongs to the renin-angiotensin system (RAS). RAS is an endocrine system that has been classically associated with regulating blood pressure and fluid and electrolyte balance; however, it is also involved in promoting inflammation, proliferation, fibrogenesis, and lipogenesis. Two pathways constitute the RAS with counter-balancing effects, which is the key to its regulation. The first axis (classical) is composed of angiotensin-converting enzyme (ACE), angiotensin (Ang) II, and angiotensin type 1 receptor (AT1R) as the main effector, which -when activated- increases the production of aldosterone and antidiuretic hormone, sympathetic nervous system tone, blood pressure, vasoconstriction, fibrosis, inflammation, and reactive oxygen species (ROS) production. Both systemic and local classical RAS' within the brain are associated with cognitive impairment, cell death, and inflammation. The second axis (non-classical or alternative) includes ACE2, which converts Ang II to Ang-(1-7), a peptide molecule that activates Mas receptor (MasR) in charge of opposing Ang II/AT1R actions. Thus, the alternative RAS axis enhances cognition, synaptic remodeling, cell survival, cell signal transmission, and antioxidant/anti-inflammatory mechanisms in the brain. In a physiological state, both RAS axes remain balanced. However, some factors can dysregulate systemic and local RAS arms. The binding of SARS-CoV-2 to ACE2 causes the internalization and degradation of this enzyme, reducing its activity, and disrupting the balance of systemic and local RAS, which partially explain the appearance of some of the neurological symptoms associated with COVID-19. Therefore, this review aims to analyze the role of RAS in the development of the neurological effects due to SARS-CoV-2 infection. Moreover, we will discuss the RAS-molecular targets that could be used for therapeutic purposes to treat the short and long-term neurological COVID-19-related sequelae.
Collapse
Affiliation(s)
- Lucía A. Méndez-García
- Laboratory of Immunometabolism, Research Division, General Hospital of Mexico “Dr. Eduardo Liceaga,”Mexico City, Mexico
| | - Galileo Escobedo
- Laboratory of Immunometabolism, Research Division, General Hospital of Mexico “Dr. Eduardo Liceaga,”Mexico City, Mexico
| | - Alan Gerardo Minguer-Uribe
- Laboratory of Molecular Neuropathology, Cellular Physiology Institute, National Autonomous University of Mexico, Mexico City, Mexico
| | - Rebeca Viurcos-Sanabria
- Laboratory of Immunometabolism, Research Division, General Hospital of Mexico “Dr. Eduardo Liceaga,”Mexico City, Mexico
- PECEM, School of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| | - José A. Aguayo-Guerrero
- Laboratory of Immunometabolism, Research Division, General Hospital of Mexico “Dr. Eduardo Liceaga,”Mexico City, Mexico
| | - José Damián Carrillo-Ruiz
- Research Directorate, General Hospital of Mexico “Dr. Eduardo Liceaga,”Mexico City, Mexico
- Department of Neurology and Neurosurgery, General Hospital of Mexico “Dr. Eduardo Liceaga,”Mexico City, Mexico
- Facultad de Ciencias de la Salud, Universidad Anáhuac, Huixquilucan, Mexico
| | | |
Collapse
|
6
|
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
|
7
|
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
|
8
|
Zhao W, Zhao YL, Liu M, Liu L, Wang Y. Possible repair mechanisms of renin-angiotensin system inhibitors, matrix metalloproteinase-9 inhibitors and protein hormones on methamphetamine-induced neurotoxicity. Mol Biol Rep 2021; 48:7509-7516. [PMID: 34623593 DOI: 10.1007/s11033-021-06741-y] [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: 06/12/2021] [Accepted: 09/08/2021] [Indexed: 12/31/2022]
Abstract
Methamphetamine is a highly addictive central stimulant with extensive and strong neurotoxicity. The neurotoxicity of methamphetamine is closely related to the imbalance of dopamine levels and the destruction of the blood-brain barrier. An increase in dopamine may induce adverse effects such as behavioral sensitization and excessive locomotion. Damage to the blood-brain barrier can cause toxic or harmful substances to leak to the central nervous system, leading to neurotoxicity. The renin-angiotensin system is essential for the regulation of dopamine levels in the brain. Matrix metalloproteinase-9 causes reward effects and behavioral sensitization by inducing dopamine release. Prolactin has been shown to be involved in the regulation of tight junction proteins and the integrity of the blood-brain barrier. At present, the treatment of methamphetamine detoxification is still based on psychotherapy, and there is no specific medicine. With the rapid increase in global seizures of methamphetamine, the treatment of its toxicity has attracted more and more attention. This review intends to summarize the therapeutic mechanisms of renin-angiotensin inhibitors, matrix metalloproteinase-9 inhibitors and protein hormones (prolactin) on methamphetamine neurotoxicity. The repair effects of these three on methamphetamine may be related to the maintenance of brain dopamine balance and the integrity of the blood-brain barrier. This review is expected to provide the new therapeutic strategy of methamphetamine toxicity.
Collapse
Affiliation(s)
- Wei Zhao
- Department of Drug Control, Criminal Investigation Police University of China, Shenyang, 110854, Liaoning, People's Republic of China.,Department of Clinical Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, People's Republic of China
| | - Yuan-Ling Zhao
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, People's Republic of China
| | - Ming Liu
- Department of Drug Control, Criminal Investigation Police University of China, Shenyang, 110854, Liaoning, People's Republic of China
| | - Lian Liu
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, People's Republic of China
| | - Yun Wang
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, People's Republic of China.
| |
Collapse
|
9
|
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
|
10
|
Wright JW, Church KJ, Harding JW. Hepatocyte Growth Factor and Macrophage-stimulating Protein "Hinge" Analogs to Treat Pancreatic Cancer. Curr Cancer Drug Targets 2020; 19:782-795. [PMID: 30914029 DOI: 10.2174/1568009619666190326130008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 03/18/2019] [Accepted: 03/20/2019] [Indexed: 12/20/2022]
Abstract
Pancreatic cancer (PC) ranks twelfth in frequency of diagnosis but is the fourth leading cause of cancer related deaths with a 5 year survival rate of less than 7 percent. This poor prognosis occurs because the early stages of PC are often asymptomatic. Over-expression of several growth factors, most notably vascular endothelial growth factor (VEGF), has been implicated in PC resulting in dysfunctional signal transduction pathways and the facilitation of tumor growth, invasion and metastasis. Hepatocyte growth factor (HGF) acts via the Met receptor and has also received research attention with ongoing efforts to develop treatments to block the Met receptor and its signal transduction pathways. Macrophage-stimulating protein (MSP), and its receptor Ron, is also recognized as important in the etiology of PC but is less well studied. Although the angiotensin II (AngII)/AT1 receptor system is best known for mediating blood pressure and body water/electrolyte balance, it also facilitates tumor vascularization and growth by stimulating the expression of VEGF. A metabolite of AngII, angiotensin IV (AngIV) has sequence homology with the "hinge regions" of HGF and MSP, key structures in the growth factor dimerization processes necessary for Met and Ron receptor activation. We have developed AngIV-based analogs designed to block dimerization of HGF and MSP and thus receptor activation. Norleual has shown promise as tested utilizing PC cell cultures. Results indicate that cell migration, invasion, and pro-survival functions were suppressed by this analog and tumor growth was significantly inhibited in an orthotopic PC mouse model.
Collapse
Affiliation(s)
- John W Wright
- Department of Psychology, Washington State University, Pullman, WA, United States.,Department of Integrative Physiology and Neuroscience, and Program in Biotechnology, Washington State University, Pullman, WA, United States
| | - Kevin J Church
- Department of Integrative Physiology and Neuroscience, and Program in Biotechnology, Washington State University, Pullman, WA, United States
| | - Joseph W Harding
- Department of Psychology, Washington State University, Pullman, WA, United States.,Department of Integrative Physiology and Neuroscience, and Program in Biotechnology, Washington State University, Pullman, WA, United States
| |
Collapse
|
11
|
Royea J, Hamel E. Brain angiotensin II and angiotensin IV receptors as potential Alzheimer's disease therapeutic targets. GeroScience 2020; 42:1237-1256. [PMID: 32700176 DOI: 10.1007/s11357-020-00231-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 07/07/2020] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that is multifactorial in nature. Yet, despite being the most common form of dementia in the elderly, AD's primary cause remains unknown. As such, there is currently little to offer AD patients as the vast majority of recently tested therapies have either failed in well-controlled clinical trials or inadequately treat AD. Recently, emerging preclinical and clinical evidence has associated the brain renin angiotensin system (RAS) to AD pathology. Accordingly, various components of the brain RAS were shown to be altered in AD patients and mouse models, including the angiotensin II type 1 (AT1R), angiotensin IV receptor (AT4R), and Mas receptors. Collectively, the changes observed within the RAS have been proposed to contribute to many of the neuropathological hallmarks of AD, including the neuronal, cognitive, and vascular dysfunctions. Accumulating evidence has additionally identified antihypertensive medications targeting the RAS, particularly angiotensin receptor blockers (ARBs) and angiotensin-converting enzyme inhibitors (ACEIs), to delay AD onset and progression. In this review, we will discuss the emergence of the RAS's involvement in AD and highlight putative mechanisms of action underlying ARB's beneficial effects that may explain their ability to modify the risk of developing AD or AD progression. The RAS may provide novel molecular targets for recovering memory pathways, cerebrovascular function, and other pathological landmarks of AD.
Collapse
Affiliation(s)
- Jessika Royea
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, 3801 University Street, Montréal, QC, H3A 2B4, Canada
| | - Edith Hamel
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, 3801 University Street, Montréal, QC, H3A 2B4, Canada.
| |
Collapse
|
12
|
Khaliq OP, Konoshita T, Moodley J, Naicker T. Soluble angiotensin IV receptor levels in preeclampsia: is there a variation? J Matern Fetal Neonatal Med 2020; 35:1156-1161. [PMID: 32208780 DOI: 10.1080/14767058.2020.1743665] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Objective: To measure the concentration of plasma soluble angiotensin IV receptor (sAT-4), a component of the renin-angiotensin system in healthy normotensive pregnancies and preeclampsia.Study design: Stored maternal plasma samples obtained at the time of diagnosis from pregnant women of African ancestry were stratified into normotensive and preeclampsia groups. Preeclampsia was subdivided into early-onset, late-onset, and into and severe preeclampsia. Plasma concentrations of sAT-4 were measured at 450 nm using the ELISA technique (LNPEP KIT).Results: The systolic and diastolic blood pressure (BP) levels of the normotensive group were statistically lower compared to preeclampsia groups (p < .05) and the mean gestational age in early-onset preeclampsia was lower compared to late-onset preeclampsia and the normotensive group (p < .05). Plasma sAT-4 levels were significantly elevated (p < .0001) in the normotensive group (median 1.95, range 1.89-2.02 ng/ml) compared to the preeclampsia group (median 1.55, range 1.42-1.74 ng/ml), regardless of gestational age. Soluble AT-4 was decreased in relation to the severity of preeclampsia (p < .001), the level in preeclampsia without severe features (median 1.57, range 1.42-1.74 ng/ml) was significantly higher than in preeclampsia with severe features (median 1.51, range 1.42-1.55 ng/ml). There was no significant difference in the sAT-4 level between early-onset preeclampsia (1.60 ± 0.13 ng/ml) and late-onset preeclampsia (1.65 ± 0.29 ng/ml) groups (p = .59).Conclusion: Plasma circulating levels of sAT-4 in women with severe features of preeclampsia had lower levels than normotensives and those with preeclampsia without severe features.
Collapse
Affiliation(s)
- Olive P Khaliq
- Optics and Imaging Centre, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Tadashi Konoshita
- Third Department of Internal Medicine, University of Fukui Faculty of Medicine Sciences, Fukui, Japan
| | - Jagidesa Moodley
- Department of Obstetrics and Gynecology and Women's Health and HIV Research Group, Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Thajasvarie Naicker
- Optics and Imaging Centre, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
13
|
Souza LAC, Trebak F, Kumar V, Satou R, Kehoe PG, Yang W, Wharton W, Feng Earley Y. Elevated cerebrospinal fluid sodium in hypertensive human subjects with a family history of Alzheimer's disease. Physiol Genomics 2020; 52:133-142. [PMID: 31961762 DOI: 10.1152/physiolgenomics.00093.2019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
High salt (sodium) intake leads to the development of hypertension despite the fact that plasma sodium concentration ([Na+]) is usually normal in hypertensive human patients. Increased cerebrospinal fluid (CSF) sodium contributes to elevated sympathetic activity and high blood pressure (BP) in rodent models of hypertension. However, whether there is an increased accumulation of sodium in the CSF of humans with chronic hypertension is not well defined. Here, we investigated CSF [Na+] from hypertensive and normotensive human subjects with family histories of Alzheimer's disease in samples collected in a clinical trial, as spinal tap is not a routine clinical procedure for hypertensive patients. The [Na+] and osmolality in plasma and CSF were measured by flame photometry. Daytime ambulatory BP was monitored while individuals were awake. Participants were deidentified and data were analyzed in conjunction with a retrospective analysis of patient history and diagnosis. We found that CSF [Na+] was significantly higher in participants with high BP compared with normotensive participants; there was no difference in plasma [Na+], or plasma and CSF osmolality between groups. Subsequent multiple linear regression analyses controlling for age, sex, race, and body mass index revealed a significant positive correlation between CSF [Na+] and BP but showed no correlation between plasma [Na+] and BP. In sum, CSF [Na+] was higher in chronic hypertensive individuals and may play a key role in the pathogenesis of human hypertension. Collectively, our findings provide evidence for the clinical significance of CSF [Na+] in chronic hypertension in humans.
Collapse
Affiliation(s)
- Lucas A C Souza
- Departments of Pharmacology and Physiology & Cell Biology, University of Nevada, Reno, School of Medicine, Reno, Nevada.,Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada
| | - Fatima Trebak
- Departments of Pharmacology and Physiology & Cell Biology, University of Nevada, Reno, School of Medicine, Reno, Nevada.,Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada
| | - Veena Kumar
- Department of Neurology, Emory University School of Nursing, Atlanta, Georgia
| | - Ryousuke Satou
- Department of Physiology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Patrick G Kehoe
- Institute of Clinical Neurosciences, University of Bristol, Bristol, United Kingdom
| | - Wei Yang
- University of Nevada, Reno, School of Community Health Sciences, Reno, Nevada
| | - Whitney Wharton
- Department of Neurology, Emory University School of Nursing, Atlanta, Georgia
| | - Yumei Feng Earley
- Departments of Pharmacology and Physiology & Cell Biology, University of Nevada, Reno, School of Medicine, Reno, Nevada.,Center for Molecular and Cellular Signaling in the Cardiovascular System, University of Nevada, Reno, Reno, Nevada
| |
Collapse
|
14
|
Royea J, Martinot P, Hamel E. Memory and cerebrovascular deficits recovered following angiotensin IV intervention in a mouse model of Alzheimer's disease. Neurobiol Dis 2019; 134:104644. [PMID: 31669735 DOI: 10.1016/j.nbd.2019.104644] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 10/01/2019] [Accepted: 10/16/2019] [Indexed: 10/25/2022] Open
Abstract
Angiotensin II type 1 receptor antagonists like losartan have been found to lower the incidence and progression to Alzheimer's disease (AD), as well as rescue cognitive and cerebrovascular deficits in AD mouse models. We previously found that co-administration of an angiotensin IV (AngIV) receptor (AT4R) antagonist prevented losartan's benefits, identifying AT4Rs as a possible target to counter AD pathogenesis. Therein, we investigated whether directly targeting AT4Rs could counter AD pathogenesis in a well-characterized mouse model of AD. Wild-type and human amyloid precursor protein (APP) transgenic (J20 line) mice (4.5 months old) received vehicle or AngIV (~1.3 nmol/day, 1 month) intracerebroventricularly via osmotic minipumps. AngIV restored short-term memory, spatial learning and memory in APP mice. AngIV normalized hippocampal AT4R levels, increased hippocampal subgranular zone cellular proliferation and dendritic arborization, and reduced oxidative stress. AngIV rescued whisker-evoked neurovascular coupling, endothelial- and smooth muscle cell-mediated cerebral vasodilatory responses, and cerebrovascular nitric oxide bioavailability. AngIV did not alter blood pressure, neuroinflammation or amyloid-β (Aβ) pathology. These preclinical findings identify AT4R as a promising target to counter Aβ-related cognitive and cerebrovascular deficits in AD.
Collapse
Affiliation(s)
- Jessika Royea
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada
| | - Pauline Martinot
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada
| | - Edith Hamel
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada.
| |
Collapse
|
15
|
Fouda AY, Fagan SC, Ergul A. Brain Vasculature and Cognition. Arterioscler Thromb Vasc Biol 2019; 39:593-602. [PMID: 30816798 PMCID: PMC6540805 DOI: 10.1161/atvbaha.118.311906] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 02/15/2019] [Indexed: 12/18/2022]
Abstract
There is a complex interaction between the brain and the cerebral vasculature to meet the metabolic demands of the brain for proper function. Preservation of cerebrovascular function and integrity has a central role in this sophisticated communication within the brain, and any derangements can have deleterious acute and chronic consequences. In almost all forms of cognitive impairment, from mild to Alzheimer disease, there are changes in cerebrovascular function and structure leading to decreased cerebral blood flow, which may initiate or worsen cognitive impairment. In this focused review, we discuss the contribution of 2 major vasoactive pathways to cerebrovascular dysfunction and cognitive impairment in an effort to identify early intervention strategies.
Collapse
Affiliation(s)
- Abdelrahman Y. Fouda
- Vascular Biology Center, Augusta University, GA
- Charlie Norwood VA Medical Center Augusta, GA
| | - Susan C. Fagan
- Program in Clinical and Experimental Therapeutics, University of Georgia College of Pharmacy, GA
- Charlie Norwood VA Medical Center Augusta, GA
| | - Adviye Ergul
- Ralph Johnson Veterans Administration Medical Center, Medical University of South Carolina, Charleston, SC
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC
| |
Collapse
|
16
|
Wang QG, Xue X, Yang Y, Gong PY, Jiang T, Zhang YD. Angiotensin IV suppresses inflammation in the brains of rats with chronic cerebral hypoperfusion. J Renin Angiotensin Aldosterone Syst 2019; 19:1470320318799587. [PMID: 30223703 PMCID: PMC6144503 DOI: 10.1177/1470320318799587] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
INTRODUCTION This study aimed to evaluate the influence of central angiotensin IV (Ang IV) infusion on chronic cerebral hypoperfusion (CCH)-related neuropathological changes including amyloid-β (Aβ), hyperphosphorylated tau (p-tau) and the inflammatory response. MATERIALS AND METHODS Rats with CCH received central infusion of Ang IV, its receptor AT4R antagonist divalinal-Ang IV or artificial cerebrospinal fluid for six weeks. During this procedure, the systolic blood pressure (SBP) was monitored, and the levels of Aβ42, p-tau and pro-inflammatory cytokines in the brain were detected. RESULTS Rats with CCH exhibited higher levels of Aβ42, p-tau and pro-inflammatory cytokines in the brain when compared with controls. Infusion of Ang IV significantly reduced the expression of pro-inflammatory cytokines in the brains of rats with CCH. Meanwhile, the reduction of pro-inflammatory cytokines levels caused by Ang IV was reversed by divalinal-Ang IV. During the treatment, the SBP in rats was not significantly altered. CONCLUSION This study demonstrates for the first time that Ang IV dose-dependently suppresses inflammation through AT4R in the brains of rats with CCH, which is independent from SBP. These findings suggest that Ang IV/AT4R may represent a potential therapeutic target for CCH-related neurological diseases.
Collapse
Affiliation(s)
- Qing-Guang Wang
- 1 Department of Neurology, Nanjing First Hospital, Nanjing Medical University, People's Republic of China.,2 Department of Neurology, Jiangyin People's Hospital, Nanjing Medical University, People's Republic of China
| | - Xiao Xue
- 1 Department of Neurology, Nanjing First Hospital, Nanjing Medical University, People's Republic of China
| | - Yang Yang
- 1 Department of Neurology, Nanjing First Hospital, Nanjing Medical University, People's Republic of China
| | - Peng-Yu Gong
- 1 Department of Neurology, Nanjing First Hospital, Nanjing Medical University, People's Republic of China
| | - Teng Jiang
- 1 Department of Neurology, Nanjing First Hospital, Nanjing Medical University, People's Republic of China
| | - Ying-Dong Zhang
- 1 Department of Neurology, Nanjing First Hospital, Nanjing Medical University, People's Republic of China
| |
Collapse
|
17
|
Shevela E, Davydova M, Starostina N, Yankovskaya A, Ostanin A, Chernykh E. Intranasal delivery of M2 macrophage-derived soluble products reduces neuropsychological deficit in patients with cerebrovascular disease: a pilot study. JOURNAL OF NEURORESTORATOLOGY 2019. [DOI: 10.26599/jnr.2019.9040010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Objective: We assessed the safety and clinical effectiveness of intranasal therapy with M2 macrophage-derived soluble products (M2-SPs) for treating patients with cerebrovascular disease (CVD). Materials and methods: The protocol of the study was registered at www.ClinicalTrails.gov (NCT02957123). The study group comprised 30 patients with chronic CVD. Neurological status was examined before therapy and at 1- and 6-month follow–up. Concentrations of 32 cytokines in the blood serum were evaluated before and 1 month after therapy onset. Neurological assessment was conducted with the following scales: Subjective Assessment of Clinical (neurological) Symptoms (SACS), Hospital Anxiety and Depression Scale (HADS), Functional Mobility Assessment in Eldery Patients (FMA), and Montreal Cognitive Assessment (MoCa). Results: M2-SPs treatment (once daily for 28~30 days) was found to be safe and well tolerated. Neuropsychological improvements showed the amelioration of neurological symptoms, reduction in anxiety and depression levels, improvement in balance and gait ability as well as cognitive functions. Clinical effects could be detected at the end of treatment course and was stable during 6-month follow-up. Blood serum cytokine evaluation demonstrated diminished baseline levels of many cytokines including those with neurotrophic activity (brain-derived neurotrophic factor, BDNF; hepatocyte growth factor, HGF; migration inhibitory factor, MIF). Upon treatment, most pronounced clinical responses were observed in patients with most severe cytokine deficiency and post-therapy normalization of MIF and HGF levels. Conclusion: Intranasal therapy with M2-SPs is safe and according to preliminary data reduces neuropsychological deficit in patients with chronic CVD. The positive effect of M2-SPs treatment seems to be HGF- and MIF-dependent.
Collapse
|
18
|
Abrahão MV, Dos Santos NFT, Kuwabara WMT, do Amaral FG, do Carmo Buonfiglio D, Peres R, Vendrame RFA, Flávio da Silveira P, Cipolla-Neto J, Baltatu OC, Afeche SC. Identification of insulin-regulated aminopeptidase (IRAP) in the rat pineal gland and the modulation of melatonin synthesis by angiotensin IV. Brain Res 2018; 1704:40-46. [PMID: 30222958 DOI: 10.1016/j.brainres.2018.09.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 08/29/2018] [Accepted: 09/11/2018] [Indexed: 02/06/2023]
Abstract
A local renin-angiotensin system (RAS) has been postulated in the pineal gland. In addition to angiotensin II (Ang II), other active metabolites have been described. In this study, we aimed to investigate a role for Ang IV in melatonin synthesis and the presence of its proposed (IRAP)/AT4 receptor (insulin-regulated aminopeptidase) in the pineal gland. The effect of Ang IV on melatonin synthesis was investigated in vitro using isolated pinealocytes. IRAP protein expression and activity were evaluated by Western blot and fluorimetry using Leu-4Me-β-naphthylamide as a substrate. Melatonin was analyzed by HPLC, calcium content by confocal microscopy and cAMP by immunoassay. Ang IV significantly augmented the NE-induced melatonin synthesis to a similar degree as that achieved by Ang II. This Ang IV effect in pinealocytes appears to be mediated by an increase in the intracellular calcium content but not by cAMP. The (IRAP)/AT4 expression and activity were identified in the pineal gland, which were significantly higher in membrane fractions than in soluble fractions. Ang IV significantly reduced IRAP activity in the pineal membrane fractions. The main findings of the present study are as follows: (1) Ang IV potentiates NE-stimulated melatonin production in pinealocytes, (2) the (IRAP)/AT4 receptor is present in the rat pineal gland, and (3) Ang IV inhibits IRAP activity and increases pinealocytes [Ca2+]i. We conclude that Ang IV is an important component of RAS and modulates melatonin synthesis in the rat pineal gland.
Collapse
Affiliation(s)
| | | | - Wilson Mitsuo Tatagiba Kuwabara
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, 05508-900 São Paulo, SP, Brazil
| | - Fernanda Gaspar do Amaral
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, 05508-900 São Paulo, SP, Brazil; Department of Physiology, Federal University of São Paulo, 04023-901 São Paulo, SP, Brazil
| | - Daniella do Carmo Buonfiglio
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, 05508-900 São Paulo, SP, Brazil
| | - Rafael Peres
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, 05508-900 São Paulo, SP, Brazil
| | | | | | - José Cipolla-Neto
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, 05508-900 São Paulo, SP, Brazil
| | - Ovidiu Constantin Baltatu
- Center of Innovation, Technology and Education (CITE), Anhembi Morumbi University-Laureate International Universities, 12247-016 São José dos Campos, SP, Brazil
| | | |
Collapse
|
19
|
Jiang L, Zhu R, Bu Q, Li Y, Shao X, Gu H, Kong J, Luo L, Long H, Guo W, Tian J, Zhao Y, Cen X. Brain Renin-Angiotensin System Blockade Attenuates Methamphetamine-Induced Hyperlocomotion and Neurotoxicity. Neurotherapeutics 2018; 15:500-510. [PMID: 29464572 PMCID: PMC5935642 DOI: 10.1007/s13311-018-0613-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Methamphetamine (METH) abuse has become a major public health concern worldwide without approved pharmacotherapies. The brain renin-angiotensin system (RAS) is involved in the regulation of neuronal function as well as neurological disorders. Angiotensin II (Ang II), which interacts with Ang II type 1 receptor (AT1-R) in the brain, plays an important role as a neuromodulator in dopaminergic transmission. However, the role of brain RAS in METH-induced behavior is largely unknown. Here, we revealed that repeated METH administration significantly upregulated the expression of AT1-R in the striatum of mice, but downregulated dopamine D3 receptor (D3R) expression. A specific AT1-R blocker telmisartan, which can penetrate the brain-blood barrier (BBB), or genetic deletion of AT1-R was sufficient to attenuate METH-triggered hyperlocomotion in mice. However, intraperitoneal injection of AT1-R blocker losartan, which cannot penetrate BBB, failed to attenuate METH-induced behavior. Moreover, intra-striatum re-expression of AT1 with lentiviral virus expressing AT1 reversed the weakened locomotor activity of AT1-/- mice treated with METH. Losartan alleviated METH-induced cytotoxicity in SH-SY5Y cells in vitro, which was accompanied by upregulated expressions of D3R and dopamine transporter. In addition, intraperitoneal injection of perindopril, which is a specific ACE inhibitor and can penetrate BBB, significantly attenuated METH-induced hyperlocomotor activity. Collectively, our results show that blockade of brain RAS attenuates METH-induced hyperlocomotion and neurotoxicity possibly through modulation of D3R expression. Our findings reveal a novel role of Ang II-AT1-R in METH-induced hyperlocomotion.
Collapse
Affiliation(s)
- Linhong Jiang
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, #1 Keyuan Road 4, Gaopeng Street, High-tech Development Zone, Chengdu, 610041, China
| | - Ruiming Zhu
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, #1 Keyuan Road 4, Gaopeng Street, High-tech Development Zone, Chengdu, 610041, China
| | - Qian Bu
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, #1 Keyuan Road 4, Gaopeng Street, High-tech Development Zone, Chengdu, 610041, China
- Department of Food Science and Technology, College of Light Industry, Textile and Food Engineering, Sichuan University, Chengdu, 610065, China
| | - Yan Li
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, #1 Keyuan Road 4, Gaopeng Street, High-tech Development Zone, Chengdu, 610041, China
| | - Xue Shao
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, #1 Keyuan Road 4, Gaopeng Street, High-tech Development Zone, Chengdu, 610041, China
| | - Hui Gu
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, #1 Keyuan Road 4, Gaopeng Street, High-tech Development Zone, Chengdu, 610041, China
| | - Jueying Kong
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, #1 Keyuan Road 4, Gaopeng Street, High-tech Development Zone, Chengdu, 610041, China
| | - Li Luo
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, #1 Keyuan Road 4, Gaopeng Street, High-tech Development Zone, Chengdu, 610041, China
| | - Hailei Long
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, #1 Keyuan Road 4, Gaopeng Street, High-tech Development Zone, Chengdu, 610041, China
| | - Wei Guo
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, #1 Keyuan Road 4, Gaopeng Street, High-tech Development Zone, Chengdu, 610041, China
- School of Pharmacy, Yantai University, Yantai, 264003, China
- State Key Laboratory of Long-Acting and Targeting Drug Delivery Technologies, Yantai, 264003, China
| | - Jingwei Tian
- School of Pharmacy, Yantai University, Yantai, 264003, China
- State Key Laboratory of Long-Acting and Targeting Drug Delivery Technologies, Yantai, 264003, China
| | - Yinglan Zhao
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, #1 Keyuan Road 4, Gaopeng Street, High-tech Development Zone, Chengdu, 610041, China
| | - Xiaobo Cen
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, #1 Keyuan Road 4, Gaopeng Street, High-tech Development Zone, Chengdu, 610041, China.
| |
Collapse
|
20
|
Jackson L, Eldahshan W, Fagan SC, Ergul A. Within the Brain: The Renin Angiotensin System. Int J Mol Sci 2018; 19:E876. [PMID: 29543776 PMCID: PMC5877737 DOI: 10.3390/ijms19030876] [Citation(s) in RCA: 208] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 03/10/2018] [Accepted: 03/11/2018] [Indexed: 02/07/2023] Open
Abstract
For many years, modulators of the renin angiotensin system (RAS) have been trusted by clinicians for the control of essential hypertension. It was recently demonstrated that these modulators have other pleiotropic properties independent of their hypotensive effects, such as enhancement of cognition. Within the brain, different components of the RAS have been extensively studied in the context of neuroprotection and cognition. Interestingly, a crosstalk between the RAS and other systems such as cholinergic, dopaminergic and adrenergic systems have been demonstrated. In this review, the preclinical and clinical evidence for the impact of RAS modulators on cognitive impairment of multiple etiologies will be discussed. In addition, the expression and function of different receptor subtypes within the RAS such as: Angiotensin II type I receptor (AT1R), Angiotensin II type II receptor (AT2R), Angiotensin IV receptor (AT4R), Mas receptor (MasR), and Mas-related-G protein-coupled receptor (MrgD), on different cell types within the brain will be presented. We aim to direct the attention of the scientific community to the plethora of evidence on the importance of the RAS on cognition and to the different disease conditions in which these agents can be beneficial.
Collapse
Affiliation(s)
- LaDonya Jackson
- Program in Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA.
| | - Wael Eldahshan
- Program in Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA.
| | - Susan C Fagan
- Program in Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA.
- Department of Neurology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
| | - Adviye Ergul
- Program in Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA.
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
- Charlie Norwood Veterans Affairs Medical Center, Augusta, GA 30904, USA.
| |
Collapse
|
21
|
Perez-Lloret S, Otero-Losada M, Toblli JE, Capani F. Renin-angiotensin system as a potential target for new therapeutic approaches in Parkinson's disease. Expert Opin Investig Drugs 2017; 26:1163-1173. [PMID: 28836869 DOI: 10.1080/13543784.2017.1371133] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Currently, available therapies for Parkinson's disease (PD) are symptomatic. Therefore, the search for neuroprotective drugs remains a top priority. Areas covered: In this review, the potential symptomatic or disease-modifying effect of drugs targeting the Renin-Angiotensin System (RAS) in PD will be explored. Expert opinion: The importance of nigrostriatal local RAS has only begun to be unraveled in the last decades. On one hand, there is a complex feedback cycle between RAS and dopamine (DA). On the other hand, RAS affects dopaminergic neurons vulnerability. Neuroprotective effects in animal PD models have been shown for the angiotensin-converting enzyme (ACE) inhibitors captopril and perindopril, and the AT1 receptor antagonists losartan, candesartan and telmisartan. These effects appear to be mediated by a reduction in the overproduction of reactive oxygen species. In a proof-of-concept, randomized, double-blind, crossover study in PD patients, perindopril enhanced the effect of levodopa without inducing dyskinesias. There has not been any clinical trial exploring the neuroprotective effect of RAS drugs, but one cohort study in hypertensive patients suggested a protective effect of ACE inhibitors on PD risk. RAS is a promising target for symptomatic and neuroprotective therapies in PD. Further studies in PD animal models and patients are warranted.
Collapse
Affiliation(s)
- Santiago Perez-Lloret
- a Institute of Cardiology Research , University of Buenos Aires, National Research Council (ININCA-UBA-CONICET) , Buenos Aires , Argentina
| | - Matilde Otero-Losada
- a Institute of Cardiology Research , University of Buenos Aires, National Research Council (ININCA-UBA-CONICET) , Buenos Aires , Argentina
| | - Jorge E Toblli
- a Institute of Cardiology Research , University of Buenos Aires, National Research Council (ININCA-UBA-CONICET) , Buenos Aires , Argentina
| | - Francisco Capani
- a Institute of Cardiology Research , University of Buenos Aires, National Research Council (ININCA-UBA-CONICET) , Buenos Aires , Argentina.,b Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud , Universidad Autónoma de Chile , Santiago de Chile , Chile
| |
Collapse
|
22
|
Haithem H, Ons A, Salma N, Jihène R, Mariam A, Mariem M, Mariem N, Nabila BR, Asma O, Sana BA, Sofien B, Ali B. Association between dementia and vascular disease-associated polymorphisms in a Tunisian population. Int J Neurosci 2017; 128:32-41. [PMID: 28657841 DOI: 10.1080/00207454.2017.1348353] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
PURPOSE Dementia is a multifactorial idiopathic pathology caused by clinical, eDementia is a multifactorial idiopathic pathology caused by clinical, environmental and genetic factors. Hence, its etiology is still unknown. We aimed to evaluate the association between five genetic risk factors for vascular diseases and dementia individually and when gathered in haplotypes. MATERIALS AND METHOD We enrolled 200 dementia patients and 300 controls. All subjects were genotyped for vascular diseaseassociated polymorphisms in the genes coding for Apolipoprotein-E (ApoE), angiotensin converting enzyme (ACE) and Paraoxonase-1 (PON1). RESULTS The association between dementia risk and all the studied polymorphisms except of PON1-Q192R was found to be significant. Carrying the ApoE e4 allele seems to increase dementia risk by 4.32 fold (p = 0.001). The risk associated with ACE I and PON1-L55M T alleles were lower (2.58 and 2.11 fold, p < 0.001 and p = 0.015, respectively). When combined in haplotypes, these polymorphisms showed a cumulative and synergetic effect. GTICC haplotype appears to be associated with 9-fold dementia risk (p < 0.001), whereas AADTT seems to reduce dementia risk by 80% (p = 0.003). CONCLUSION Our results suggest that, ApoE ε4, ACE I and PON1-L55M T alleles are associated with dementia risk whether these polymorphisms were studied separately or gathered in haplotypes. Still, the contribution of each gene to the pathophysiological development of dementia must be more investigated.
Collapse
Affiliation(s)
- Hamdouni Haithem
- a Biochemistry Department , Sahloul University Hospital , Sousse , Tunisia.,d Faculty of Pharmacy , University of Monastir , Monastir , Tunisia
| | - Achour Ons
- a Biochemistry Department , Sahloul University Hospital , Sousse , Tunisia.,d Faculty of Pharmacy , University of Monastir , Monastir , Tunisia
| | - Naija Salma
- b Neurology Department , Sahloul University Hospital , Sousse , Tunisia
| | - Rejeb Jihène
- a Biochemistry Department , Sahloul University Hospital , Sousse , Tunisia
| | - Aounallah Mariam
- a Biochemistry Department , Sahloul University Hospital , Sousse , Tunisia
| | - Mhiri Mariem
- b Neurology Department , Sahloul University Hospital , Sousse , Tunisia
| | - Noureddine Mariem
- a Biochemistry Department , Sahloul University Hospital , Sousse , Tunisia.,d Faculty of Pharmacy , University of Monastir , Monastir , Tunisia
| | - Ben Rejeb Nabila
- a Biochemistry Department , Sahloul University Hospital , Sousse , Tunisia.,c Biochemistry Department , Sahloul University Hospital , Sousse , Tunisia.,d Faculty of Pharmacy , University of Monastir , Monastir , Tunisia
| | - Omezzine Asma
- a Biochemistry Department , Sahloul University Hospital , Sousse , Tunisia.,c Biochemistry Department , Sahloul University Hospital , Sousse , Tunisia.,d Faculty of Pharmacy , University of Monastir , Monastir , Tunisia
| | - Ben Amor Sana
- b Neurology Department , Sahloul University Hospital , Sousse , Tunisia
| | - Benammou Sofien
- b Neurology Department , Sahloul University Hospital , Sousse , Tunisia
| | - Bouslama Ali
- a Biochemistry Department , Sahloul University Hospital , Sousse , Tunisia.,c Biochemistry Department , Sahloul University Hospital , Sousse , Tunisia.,d Faculty of Pharmacy , University of Monastir , Monastir , Tunisia
| |
Collapse
|
23
|
Huber G, Schuster F, Raasch W. Brain renin-angiotensin system in the pathophysiology of cardiovascular diseases. Pharmacol Res 2017; 125:72-90. [PMID: 28687340 DOI: 10.1016/j.phrs.2017.06.016] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 06/28/2017] [Accepted: 06/28/2017] [Indexed: 02/07/2023]
Abstract
Cardiovascular diseases (CVD) are among the main causes of death globally and in this context hypertension represents one of the key risk factors for developing a CVD. It is well established that the peripheral renin-angiotensin system (RAS) plays an important role in regulating blood pressure (BP). All components of the classic RAS can also be found in the brain but, in contrast to the peripheral RAS, how the endogenous RAS is involved in modulating cardiovascular effects in the brain is not fully understood yet. It is a complex system that may work differently in diverse areas of the brain and is linked to the peripheral system by the circumventricular organs (CVO), which do not have a blood brain barrier (BBB). In this review, we focus on the brain angiotensin peptides, their interactions with each other, and the consequences in the central nervous system (CNS) concerning cardiovascular control. Additionally, we present potential drug targets in the brain RAS for the treatment of hypertension.
Collapse
Affiliation(s)
- Gianna Huber
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Germany; CBBM (Center of Brain, Behavior and Metabolism), Lübeck, Germany
| | - Franziska Schuster
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Germany; CBBM (Center of Brain, Behavior and Metabolism), Lübeck, Germany
| | - Walter Raasch
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Germany; CBBM (Center of Brain, Behavior and Metabolism), Lübeck, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany.
| |
Collapse
|
24
|
Hallberg M, Sumners C, Steckelings UM, Hallberg A. Small-molecule AT2 receptor agonists. Med Res Rev 2017; 38:602-624. [DOI: 10.1002/med.21449] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 05/03/2017] [Accepted: 05/16/2017] [Indexed: 12/11/2022]
Affiliation(s)
- Mathias Hallberg
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, BMC; Uppsala University; P.O. Box 591 SE751 24 Uppsala Sweden
| | - Colin Sumners
- Department of Physiology and Functional Genomics, University of Florida; College of Medicine and McKnight Brain Institute; Gainesville FL 32611
| | - U. Muscha Steckelings
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research; University of Southern Denmark; P.O. Box 5230 Odense Denmark
| | - Anders Hallberg
- Department of Medicinal Chemistry, BMC; Uppsala University; P.O. Box 574 SE-751 23 Uppsala Sweden
| |
Collapse
|
25
|
Labandeira-Garcia JL, Rodríguez-Perez AI, Garrido-Gil P, Rodriguez-Pallares J, Lanciego JL, Guerra MJ. Brain Renin-Angiotensin System and Microglial Polarization: Implications for Aging and Neurodegeneration. Front Aging Neurosci 2017; 9:129. [PMID: 28515690 PMCID: PMC5413566 DOI: 10.3389/fnagi.2017.00129] [Citation(s) in RCA: 155] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 04/18/2017] [Indexed: 12/12/2022] Open
Abstract
Microglia can transform into proinflammatory/classically activated (M1) or anti-inflammatory/alternatively activated (M2) phenotypes following environmental signals related to physiological conditions or brain lesions. An adequate transition from the M1 (proinflammatory) to M2 (immunoregulatory) phenotype is necessary to counteract brain damage. Several factors involved in microglial polarization have already been identified. However, the effects of the brain renin-angiotensin system (RAS) on microglial polarization are less known. It is well known that there is a “classical” circulating RAS; however, a second RAS (local or tissue RAS) has been observed in many tissues, including brain. The locally formed angiotensin is involved in local pathological changes of these tissues and modulates immune cells, which are equipped with all the components of the RAS. There are also recent data showing that brain RAS plays a major role in microglial polarization. Level of microglial NADPH-oxidase (Nox) activation is a major regulator of the shift between M1/proinflammatory and M2/immunoregulatory microglial phenotypes so that Nox activation promotes the proinflammatory and inhibits the immunoregulatory phenotype. Angiotensin II (Ang II), via its type 1 receptor (AT1), is a major activator of the NADPH-oxidase complex, leading to pro-oxidative and pro-inflammatory effects. However, these effects are counteracted by a RAS opposite arm constituted by Angiotensin II/AT2 receptor signaling and Angiotensin 1–7/Mas receptor (MasR) signaling. In addition, activation of prorenin-renin receptors may contribute to activation of the proinflammatory phenotype. Aged brains showed upregulation of AT1 and downregulation of AT2 receptor expression, which may contribute to a pro-oxidative pro-inflammatory state and the increase in neuron vulnerability. Several recent studies have shown interactions between the brain RAS and different factors involved in microglial polarization, such as estrogens, Rho kinase (ROCK), insulin-like growth factor-1 (IGF-1), tumor necrosis factor α (TNF)-α, iron, peroxisome proliferator-activated receptor gamma, and toll-like receptors (TLRs). Metabolic reprogramming has recently been involved in the regulation of the neuroinflammatory response. Interestingly, we have recently observed a mitochondrial RAS, which is altered in aged brains. In conclusion, dysregulation of brain RAS plays a major role in aging-related changes and neurodegeneration by exacerbation of oxidative
stress (OS) and neuroinflammation, which may be attenuated by pharmacological manipulation of RAS components.
Collapse
Affiliation(s)
- Jose L Labandeira-Garcia
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de CompostelaSantiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED)Madrid, Spain
| | - Ana I Rodríguez-Perez
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de CompostelaSantiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED)Madrid, Spain
| | - Pablo Garrido-Gil
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de CompostelaSantiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED)Madrid, Spain
| | - Jannette Rodriguez-Pallares
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de CompostelaSantiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED)Madrid, Spain
| | - Jose L Lanciego
- Networking Research Center on Neurodegenerative Diseases (CIBERNED)Madrid, Spain.,Neurosciences Division, Center for Applied Medical Research (CIMA), University of NavarraPamplona, Spain
| | - Maria J Guerra
- Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de CompostelaSantiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED)Madrid, Spain
| |
Collapse
|
26
|
Patent Highlights October-November 2016. Pharm Pat Anal 2017; 6:53-60. [PMID: 28248128 DOI: 10.4155/ppa-2017-0001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
A snapshot of noteworthy recent developments in the patent literature of relevance to pharmaceutical and medical research and development.
Collapse
|
27
|
Dong J, Cui Y, Li S, Le W. Current Pharmaceutical Treatments and Alternative Therapies of Parkinson's Disease. Curr Neuropharmacol 2016; 14:339-55. [PMID: 26585523 PMCID: PMC4876590 DOI: 10.2174/1570159x14666151120123025] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 07/16/2015] [Accepted: 10/09/2015] [Indexed: 01/01/2023] Open
Abstract
Over the decades, pharmaceutical treatments, particularly dopaminergic (DAergic) drugs have been considered as the main therapy against motor symptoms of Parkinson's disease (PD). It is proposed that DAergic drugs in combination with other medications, such as monoamine oxidase type B inhibitors, catechol-O-methyl transferase inhibitors, anticholinergics and other newly developed non-DAergic drugs can make a better control of motor symptoms or alleviate levodopa-induced motor complications. Moreover, non-motor symptoms of PD, such as cognitive, neuropsychiatric, sleep, autonomic and sensory disturbances caused by intrinsic PD pathology or drug-induced side effects, are gaining increasing attention and urgently need to be taken care of due to their impact on quality of life. Currently, neuroprotective therapies have been investigated extensively in pre-clinical studies, and some of them have been subjected to clinical trials. Furthermore, non-pharmaceutical treatments, including deep brain stimulation (DBS), gene therapy, cell replacement therapy and some complementary managements, such as Tai chi, Yoga, traditional herbs and molecular targeted therapies have also been considered as effective alternative therapies to classical pharmaceutics. This review will provide us updated information regarding the current drugs and non-drugs therapies for PD.
Collapse
Affiliation(s)
| | | | | | - Weidong Le
- Neurology and Director of Center for Translational Research of Neurological Diseases, 1st Affiliated Hospital, Dalian Medical University, Dalian 116021, Liaoning Province, China.
| |
Collapse
|
28
|
Ongali B, Nicolakakis N, Tong XK, Aboulkassim T, Imboden H, Hamel E. Enalapril Alone or Co-Administered with Losartan Rescues Cerebrovascular Dysfunction, but not Mnemonic Deficits or Amyloidosis in a Mouse Model of Alzheimer's Disease. J Alzheimers Dis 2016; 51:1183-95. [PMID: 26923013 DOI: 10.3233/jad-150868] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The co-administration of angiotensin converting enzyme inhibitors (ACEi) and angiotensin II (AngII) receptor blockers (ARB) that bind angiotensin type 1 receptors (AT1R) may protect from Alzheimer's disease (AD) better than each treatment taken alone. We tested the curative potential of the non brain-penetrant ACEi enalapril (3 mg/kg/day) administered for 3 months either alone or in combination with the brain penetrant ARB losartan (10 mg/kg/day) in aged (∼15 months) transgenic mice overexpressing a mutated form of the human amyloid-β protein precursor (AβPP, thereafter APP mice). We studied cerebrovascular function, protein levels of oxidative stress markers (superoxide dismutases SOD1, SOD2 and the NADPH oxidase subunit p67phox), amyloid-β (Aβ) pathology, astrogliosis, cholinergic innervation, AT1R and angiotensin IV receptor (AT4R) levels, together with cognitive performance. Both treatments normalized cerebrovascular reactivity and p67phox protein levels, but they did not reduce the cerebrovascular levels of SOD1. Combined treatment normalized cerebrovascular SOD2 levels, significantly attenuated astrogliosis, but did not reduce the increased levels of cerebrovascular AT1R. Yet, combined therapy enhanced thioflavin-S labeled Aβ plaque burden, a tendency not significant when Aβ1 - 42 plaque load was considered. None of the treatments rescued cognitive deficits, cortical AT4R or cholinergic innervation. We conclude that both treatments normalized cerebrovascular function by inhibiting the AngII-induced oxidative stress cascade, and that the positive effects of the combined therapy on astrogliosis were likely due to the ability of losartan to enter brain parenchyma. However, enalapril did not potentiate, and may even dampen, the reported cognitive benefits of losartan, raising caution when selecting the most appropriate antihypertensive therapy in AD patients.
Collapse
Affiliation(s)
- Brice Ongali
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Nektaria Nicolakakis
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Xing-Kang Tong
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Tahar Aboulkassim
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Hans Imboden
- Institute of Cell Biology, University of Bern, Switzerland
| | - Edith Hamel
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, QC, Canada
| |
Collapse
|
29
|
Tetramethylpyrazine Analogue CXC195 Protects Against Dopaminergic Neuronal Apoptosis via Activation of PI3K/Akt/GSK3β Signaling Pathway in 6-OHDA-Induced Parkinson's Disease Mice. Neurochem Res 2016; 42:1141-1150. [PMID: 28005221 DOI: 10.1007/s11064-016-2148-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 12/08/2016] [Accepted: 12/10/2016] [Indexed: 01/06/2023]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder and characterized by motor system disorders resulting in loss of dopaminergic (DA) neurons. CXC195, a novel tetramethylpyrazine derivative, has been shown strongest neuroprotective effects due to its anti-apoptotic activity. However, whether CXC195 protects against DA neuronal damage in PD and the mechanisms underlying its beneficial effects are unknown. The purpose of our study was to investigate the potential neuroprotective role of CXC195 and to elucidate its mechanism of action against 6-hydroxydopamine (6-OHDA)-induced mouse model of PD. CXC195 administration improved DA neurodegeneration in PD mice induced by 6-OHDA. Our further findings confirmed treatment of CXC195 at the dose of 10 mg/kg significantly inhibited the apoptosis by decreasing the level of cleaved caspase-3 and Bax, and increasing the level of Bcl-2 in 6-OHDA-lesioned mice. Meanwhile, 6-OHDA also decreased the amount of phosphorylated Akt while increased GSK-3β activity (the amount of phosphorylated GSK-3β at Ser9 was decreased) which was prevented by CXC195. Wortmannin, a specific PI3K inhibitor, dramatically abolished the changes induced by CXC195. Our study firstly demonstrated that CXC195 protected against DA neurodegeneration in 6-OHDA-induced PD model by its anti-apoptotic properties and PI3K/Akt/GSK3β signaling pathway was involved in it.
Collapse
|
30
|
El-Hawli A, Qaradakhi T, Hayes A, Rybalka E, Smith R, Caprnda M, Opatrilova R, Gazdikova K, Benckova M, Kruzliak P, Zulli A. IRAP inhibition using HFI419 prevents moderate to severe acetylcholine mediated vasoconstriction in a rabbit model. Biomed Pharmacother 2016; 86:23-26. [PMID: 27936390 DOI: 10.1016/j.biopha.2016.11.142] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 11/26/2016] [Accepted: 11/30/2016] [Indexed: 10/20/2022] Open
Abstract
Coronary artery vasospasm (constriction) caused by reduced nitric oxide bioavailability leads to myocardial infarction. Reduced endothelial release of nitric oxide by the neurotransmitter acetylcholine, leads to paradoxical vasoconstriction as it binds to smooth muscle cell M3 receptors. Thus, inhibition of coronary artery vasospasm will improve clinical outcomes. Inhibition of insulin regulated aminopeptidase has been shown to improve vessel function, thus we tested the hypothesis that HFI419, an inhibitor of insulin regulated aminopeptidase, could reduce blood vessel constriction to acetylcholine. The abdominal aorta was excised from New Zealand white rabbits (n=15) and incubated with 3mM Hcy to induce vascular dysfunction in vitro for 1h. HFI419 was added 5min prior to assessment of vascular function by cumulative doses of acetylcholine. In some rings, vasoconstriction to acetylcholine was observed in aortic rings after pre-incubation with 3mM homocysteine. Incubation with HFI419 inhibited the vasoconstrictive response to acetylcholine, thus improving, but not normalizing, vascular function (11.5±8.9% relaxation vs 79.2±37% constriction, p<0.05). Similarly, in another group with mild vasoconstriction, HFI419 inhibited this effect (34.9±4.6% relaxation vs 11.1±5.2%, constriction, p<0.05). HFI419 had no effect on control aorta or aorta with mild aortic dysfunction. The present study shows that HFI419 prevents acetylcholine mediated vasoconstriction in dysfunctional blood vessels. HFI419 had no effect on normal vasodilation. Our results indicate a therapeutic potential of HFI419 in reducing coronary artery vasospasm.
Collapse
Affiliation(s)
- Aisha El-Hawli
- Centre for Chronic Disease (CCD), College of Health & Biomedicine, Victoria University, Melbourne, Victoria, Australia
| | - Tawar Qaradakhi
- Centre for Chronic Disease (CCD), College of Health & Biomedicine, Victoria University, Melbourne, Victoria, Australia
| | - Alan Hayes
- Centre for Chronic Disease (CCD), College of Health & Biomedicine, Victoria University, Melbourne, Victoria, Australia
| | - Emma Rybalka
- Centre for Chronic Disease (CCD), College of Health & Biomedicine, Victoria University, Melbourne, Victoria, Australia
| | - Renee Smith
- Centre for Chronic Disease (CCD), College of Health & Biomedicine, Victoria University, Melbourne, Victoria, Australia
| | - Martin Caprnda
- 2nd Department of Internal Medicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Radka Opatrilova
- Department of Chemical Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences, Brno, Czechia
| | - Katarina Gazdikova
- Department of Nutrition, Faculty of Nursing and Professional Health Studies, Slovak Medical University, Bratislava, Slovak Republic; Department of General Medicine, Faculty of Medicine, Slovak Medical University, Bratislava, Slovak Republic.
| | - Maria Benckova
- Department of Medical and Clinical Biophysics, Faculty of Medicine, Pavol Jozef Safarik University, Kosice, Slovakia
| | - Peter Kruzliak
- Department of Chemical Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences, Brno, Czechia; 2nd Department of Surgery, Center for Vascular Disease, Faculty of Medicine, Masaryk University and St. Annés University Hospital, Brno, Czechia.
| | - Anthony Zulli
- Centre for Chronic Disease (CCD), College of Health & Biomedicine, Victoria University, Melbourne, Victoria, Australia.
| |
Collapse
|
31
|
Mascolo A, Sessa M, Scavone C, De Angelis A, Vitale C, Berrino L, Rossi F, Rosano G, Capuano A. New and old roles of the peripheral and brain renin-angiotensin-aldosterone system (RAAS): Focus on cardiovascular and neurological diseases. Int J Cardiol 2016; 227:734-742. [PMID: 27823897 DOI: 10.1016/j.ijcard.2016.10.069] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 10/26/2016] [Indexed: 02/06/2023]
Abstract
It is commonly accepted that the renin-angiotensin-aldosterone system (RAAS) is a cardiovascular circulating hormonal system that plays also an important role in the modulation of several patterns in the brain. The pathway of the RAAS can be divided into two classes: the traditional pathway of RAAS, also named classic RAAS, and the non-classic RAAS. Both pathways play a role in both cardiovascular and neurological diseases through a peripheral or central control. In this regard, renewed interest is growing in the last years for the consideration that the brain RAAS could represent a new important therapeutic target to regulate not only the blood pressure via central nervous control, but also neurological diseases. However, the development of compounds able to cross the blood-brain barrier and to act on the brain RAAS is challenging, especially if the metabolic stability and the half-life are taken into consideration. To date, two drug classes (aminopeptidase type A inhibitors and angiotensin IV analogues) acting on the brain RAAS are in development in pre-clinical or clinical stages. In this article, we will present an overview of the biological functions played by peripheral and brain classic and non-classic pathways of the RAAS in several clinical conditions, focusing on the brain RAAS and on the new pharmacological targets of the RAAS.
Collapse
Affiliation(s)
- A Mascolo
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Second University of Naples, Naples, Italy.
| | - M Sessa
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Second University of Naples, Naples, Italy
| | - C Scavone
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Second University of Naples, Naples, Italy
| | - A De Angelis
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Second University of Naples, Naples, Italy
| | - C Vitale
- IRCCS San Raffaele Pisana, Rome, Italy
| | - L Berrino
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Second University of Naples, Naples, Italy
| | - F Rossi
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Second University of Naples, Naples, Italy
| | - G Rosano
- IRCCS San Raffaele Pisana, Rome, Italy; Cardiovascular and Cell Sciences Research Institute, St. George's, University of London, London, UK
| | - A Capuano
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Second University of Naples, Naples, Italy
| |
Collapse
|
32
|
Wincewicz D, Juchniewicz A, Waszkiewicz N, Braszko JJ. Angiotensin II type 1 receptor blockade by telmisartan prevents stress-induced impairment of memory via HPA axis deactivation and up-regulation of brain-derived neurotrophic factor gene expression. Pharmacol Biochem Behav 2016; 148:108-18. [PMID: 27375198 DOI: 10.1016/j.pbb.2016.06.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 06/10/2016] [Accepted: 06/29/2016] [Indexed: 01/19/2023]
Abstract
Physical and psychological aspects of chronic stress continue to be a persistent clinical problem for which new pharmacological treatment strategies are aggressively sought. By the results of our previous work it has been demonstrated that telmisartan (TLM), an angiotensin type 1 receptor (AT1) blocker (ARB) and partial agonist of peroxisome proliferator-activated receptor gamma (PPARγ), alleviates stress-induced cognitive decline. Understanding of mechanistic background of this phenomenon is hampered by both dual binding sites of TLM and limited data on the consequences of central AT1 blockade and PPARγ activation. Therefore, a critical need exists for progress in the characterization of this target for pro-cognitive drug discovery. An unusual ability of novel ARBs to exert various PPARγ binding activities is commonly being viewed as predominant over angiotensin blockade in terms of neuroprotection. Here we aimed to verify this hypothesis using an animal model of chronic psychological stress (Wistar rats restrained 2.5h daily for 21days) with simultaneous oral administration of TLM (1mg/kg), GW9662 - PPARγ receptor antagonist (0.5mg/kg), or both in combination, followed by a battery of behavioral tests (open field, elevated plus maze, inhibitory avoidance - IA, object recognition - OR), quantitative determination of serum corticosterone (CORT) and evaluation of brain-derived neurotrophic factor (BDNF) gene expression in the medial prefrontal cortex (mPFC) and hippocampus (HIP). Stressed animals displayed decreased recall of the IA behavior (p<0.001), decreased OR (p<0.001), substantial CORT increase (p<0.001) and significantly downregulated expression of BDNF in the mPFC (p<0.001), which were attenuated in rats receiving TLM and TLM+GW9662. These data indicate that procognitive effect of ARBs in stressed subjects do not result from PPAR-γ activation, but AT1 blockade and subsequent hypothalamus-pituitary-adrenal axis deactivation associated with changes in primarily cortical gene expression. This study confirms the dual activities of TLM that controls hypertension and cognition through AT1 blockade.
Collapse
Affiliation(s)
- D Wincewicz
- Department of Clinical Pharmacology, Medical University of Bialystok, Waszyngtona 15a, 15274 Bialystok, Poland; Department of Psychiatry, Medical University of Bialystok, Poland.
| | - A Juchniewicz
- Department of Clinical Molecular Biology, Medical University of Bialystok, Poland
| | - N Waszkiewicz
- Department of Psychiatry, Medical University of Bialystok, Poland
| | - J J Braszko
- Department of Clinical Pharmacology, Medical University of Bialystok, Waszyngtona 15a, 15274 Bialystok, Poland
| |
Collapse
|
33
|
Renin-angiotensin system as a potential therapeutic target in stroke and retinopathy: experimental and clinical evidence. Clin Sci (Lond) 2016; 130:221-38. [PMID: 26769658 DOI: 10.1042/cs20150350] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
As our knowledge expands, it is now clear that the renin-angiotensin (Ang) system (RAS) mediates functions other than regulating blood pressure (BP). The RAS plays a central role in the pathophysiology of different neurovascular unit disorders including stroke and retinopathy. Moreover, the beneficial actions of RAS modulation in brain and retina have been documented in experimental research, but not yet exploited clinically. The RAS is a complex system with distinct yet interconnected components. Understanding the different RAS components and their functions under brain and retinal pathological conditions is crucial to reap their benefits. The aim of the present review is to provide an experimental and clinical update on the role of RAS in the pathophysiology and treatment of stroke and retinopathy. Combining the evidence from both these disorders allows a unique opportunity to move both fields forward.
Collapse
|
34
|
Hamel E, Royea J, Ongali B, Tong XK. Neurovascular and Cognitive failure in Alzheimer's Disease: Benefits of Cardiovascular Therapy. Cell Mol Neurobiol 2016; 36:219-32. [PMID: 26993506 DOI: 10.1007/s10571-015-0285-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 10/06/2015] [Indexed: 12/19/2022]
Abstract
Alzheimer's disease (AD) is a multifactorial and multifaceted disease for which we currently have very little to offer since there is no curative therapy, with only limited disease-modifying drugs. Recent studies in AD mouse models that recapitulate the amyloid-β (Aβ) pathology converge to demonstrate that it is possible to salvage cerebrovascular function with a variety of drugs and, particularly, therapies used to treat cardiovascular diseases such as hypercholesterolemia and hypertension. These drugs can reestablish dilatory function mediated by various endothelial and smooth muscle ion channels as well as nitric oxide availability, benefits that result in normalized brain perfusion. These cerebrovascular benefits would favor brain perfusion, which may help maintain neuronal function and, possibly, delay cognitive failure. However, restoring cerebrovascular function in AD mouse models was not necessarily accompanied by rescue of cognitive deficits related to spatial learning and memory. The results with cardiovascular therapies rather suggest that drugs originally designed to treat cardiovascular diseases that concurrently restore cerebrovascular and cognitive function do so through their pleiotropic effects. Specifically, recent findings suggest that these drugs act directly on brain cells and neuronal pathways involved in memory formation, hence, working simultaneously albeit independently on neuronal and vascular targets. These findings may help select medications for patients with cardiovascular diseases at risk of developing AD with increasing age. Further, they may identify molecular targets for recovering memory pathways that bear potential for new therapeutic avenues.
Collapse
Affiliation(s)
- Edith Hamel
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, 3801 University Street, Suite 748, Montréal, QC, H3A 2B4, Canada.
| | - Jessika Royea
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, 3801 University Street, Suite 748, Montréal, QC, H3A 2B4, Canada
| | - Brice Ongali
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, 3801 University Street, Suite 748, Montréal, QC, H3A 2B4, Canada
| | - Xin-Kang Tong
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, 3801 University Street, Suite 748, Montréal, QC, H3A 2B4, Canada
| |
Collapse
|
35
|
Diwakarla S, Nylander E, Grönbladh A, Vanga SR, Khan YS, Gutiérrez-de-Terán H, Ng L, Pham V, Sävmarker J, Lundbäck T, Jenmalm-Jensen A, Andersson H, Engen K, Rosenström U, Larhed M, Åqvist J, Chai SY, Hallberg M. Binding to and Inhibition of Insulin-Regulated Aminopeptidase by Macrocyclic Disulfides Enhances Spine Density. Mol Pharmacol 2016; 89:413-24. [PMID: 26769413 DOI: 10.1124/mol.115.102533] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 01/13/2016] [Indexed: 01/28/2023] Open
Abstract
Angiotensin IV (Ang IV) and related peptide analogs, as well as nonpeptide inhibitors of insulin-regulated aminopeptidase (IRAP), have previously been shown to enhance memory and cognition in animal models. Furthermore, the endogenous IRAP substrates oxytocin and vasopressin are known to facilitate learning and memory. In this study, the two recently synthesized 13-membered macrocyclic competitive IRAP inhibitors HA08 and HA09, which were designed to mimic the N terminus of oxytocin and vasopressin, were assessed and compared based on their ability to bind to the IRAP active site, and alter dendritic spine density in rat hippocampal primary cultures. The binding modes of the IRAP inhibitors HA08, HA09, and of Ang IV in either the extended or γ-turn conformation at the C terminus to human IRAP were predicted by docking and molecular dynamics simulations. The binding free energies calculated with the linear interaction energy method, which are in excellent agreement with experimental data and simulations, have been used to explain the differences in activities of the IRAP inhibitors, both of which are structurally very similar, but differ only with regard to one stereogenic center. In addition, we show that HA08, which is 100-fold more potent than the epimer HA09, can enhance dendritic spine number and alter morphology, a process associated with memory facilitation. Therefore, HA08, one of the most potent IRAP inhibitors known today, may serve as a suitable starting point for medicinal chemistry programs aided by MD simulations aimed at discovering more drug-like cognitive enhancers acting via augmenting synaptic plasticity.
Collapse
Affiliation(s)
- Shanti Diwakarla
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence (S.D., E.N., A.G., M.H.), Department of Cell and Molecular Biology (S.R.V., Y.S.K., H.G.T., J.A.), The Beijer Laboratory, Department of Medicinal Chemistry (J.S.), Department of Medicinal Chemistry (H.A., K.E., U.R.), Science for Life Laboratory, Department of Medicinal Chemistry (M.L.), BMC, Uppsala University, Uppsala, Sweden; Chemical Biology Consortium Sweden, Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medicinal Biochemistry and Biophysics (T.L., A.J.), Karolinska Institute, Sweden; and Biomedicine Discovery Institute, Department of Physiology (L.N., V.P., S.Y.C.), Monash University, Melbourne, Australia
| | - Erik Nylander
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence (S.D., E.N., A.G., M.H.), Department of Cell and Molecular Biology (S.R.V., Y.S.K., H.G.T., J.A.), The Beijer Laboratory, Department of Medicinal Chemistry (J.S.), Department of Medicinal Chemistry (H.A., K.E., U.R.), Science for Life Laboratory, Department of Medicinal Chemistry (M.L.), BMC, Uppsala University, Uppsala, Sweden; Chemical Biology Consortium Sweden, Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medicinal Biochemistry and Biophysics (T.L., A.J.), Karolinska Institute, Sweden; and Biomedicine Discovery Institute, Department of Physiology (L.N., V.P., S.Y.C.), Monash University, Melbourne, Australia
| | - Alfhild Grönbladh
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence (S.D., E.N., A.G., M.H.), Department of Cell and Molecular Biology (S.R.V., Y.S.K., H.G.T., J.A.), The Beijer Laboratory, Department of Medicinal Chemistry (J.S.), Department of Medicinal Chemistry (H.A., K.E., U.R.), Science for Life Laboratory, Department of Medicinal Chemistry (M.L.), BMC, Uppsala University, Uppsala, Sweden; Chemical Biology Consortium Sweden, Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medicinal Biochemistry and Biophysics (T.L., A.J.), Karolinska Institute, Sweden; and Biomedicine Discovery Institute, Department of Physiology (L.N., V.P., S.Y.C.), Monash University, Melbourne, Australia
| | - Sudarsana Reddy Vanga
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence (S.D., E.N., A.G., M.H.), Department of Cell and Molecular Biology (S.R.V., Y.S.K., H.G.T., J.A.), The Beijer Laboratory, Department of Medicinal Chemistry (J.S.), Department of Medicinal Chemistry (H.A., K.E., U.R.), Science for Life Laboratory, Department of Medicinal Chemistry (M.L.), BMC, Uppsala University, Uppsala, Sweden; Chemical Biology Consortium Sweden, Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medicinal Biochemistry and Biophysics (T.L., A.J.), Karolinska Institute, Sweden; and Biomedicine Discovery Institute, Department of Physiology (L.N., V.P., S.Y.C.), Monash University, Melbourne, Australia
| | - Yasmin Shamsudin Khan
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence (S.D., E.N., A.G., M.H.), Department of Cell and Molecular Biology (S.R.V., Y.S.K., H.G.T., J.A.), The Beijer Laboratory, Department of Medicinal Chemistry (J.S.), Department of Medicinal Chemistry (H.A., K.E., U.R.), Science for Life Laboratory, Department of Medicinal Chemistry (M.L.), BMC, Uppsala University, Uppsala, Sweden; Chemical Biology Consortium Sweden, Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medicinal Biochemistry and Biophysics (T.L., A.J.), Karolinska Institute, Sweden; and Biomedicine Discovery Institute, Department of Physiology (L.N., V.P., S.Y.C.), Monash University, Melbourne, Australia
| | - Hugo Gutiérrez-de-Terán
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence (S.D., E.N., A.G., M.H.), Department of Cell and Molecular Biology (S.R.V., Y.S.K., H.G.T., J.A.), The Beijer Laboratory, Department of Medicinal Chemistry (J.S.), Department of Medicinal Chemistry (H.A., K.E., U.R.), Science for Life Laboratory, Department of Medicinal Chemistry (M.L.), BMC, Uppsala University, Uppsala, Sweden; Chemical Biology Consortium Sweden, Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medicinal Biochemistry and Biophysics (T.L., A.J.), Karolinska Institute, Sweden; and Biomedicine Discovery Institute, Department of Physiology (L.N., V.P., S.Y.C.), Monash University, Melbourne, Australia
| | - Leelee Ng
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence (S.D., E.N., A.G., M.H.), Department of Cell and Molecular Biology (S.R.V., Y.S.K., H.G.T., J.A.), The Beijer Laboratory, Department of Medicinal Chemistry (J.S.), Department of Medicinal Chemistry (H.A., K.E., U.R.), Science for Life Laboratory, Department of Medicinal Chemistry (M.L.), BMC, Uppsala University, Uppsala, Sweden; Chemical Biology Consortium Sweden, Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medicinal Biochemistry and Biophysics (T.L., A.J.), Karolinska Institute, Sweden; and Biomedicine Discovery Institute, Department of Physiology (L.N., V.P., S.Y.C.), Monash University, Melbourne, Australia
| | - Vi Pham
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence (S.D., E.N., A.G., M.H.), Department of Cell and Molecular Biology (S.R.V., Y.S.K., H.G.T., J.A.), The Beijer Laboratory, Department of Medicinal Chemistry (J.S.), Department of Medicinal Chemistry (H.A., K.E., U.R.), Science for Life Laboratory, Department of Medicinal Chemistry (M.L.), BMC, Uppsala University, Uppsala, Sweden; Chemical Biology Consortium Sweden, Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medicinal Biochemistry and Biophysics (T.L., A.J.), Karolinska Institute, Sweden; and Biomedicine Discovery Institute, Department of Physiology (L.N., V.P., S.Y.C.), Monash University, Melbourne, Australia
| | - Jonas Sävmarker
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence (S.D., E.N., A.G., M.H.), Department of Cell and Molecular Biology (S.R.V., Y.S.K., H.G.T., J.A.), The Beijer Laboratory, Department of Medicinal Chemistry (J.S.), Department of Medicinal Chemistry (H.A., K.E., U.R.), Science for Life Laboratory, Department of Medicinal Chemistry (M.L.), BMC, Uppsala University, Uppsala, Sweden; Chemical Biology Consortium Sweden, Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medicinal Biochemistry and Biophysics (T.L., A.J.), Karolinska Institute, Sweden; and Biomedicine Discovery Institute, Department of Physiology (L.N., V.P., S.Y.C.), Monash University, Melbourne, Australia
| | - Thomas Lundbäck
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence (S.D., E.N., A.G., M.H.), Department of Cell and Molecular Biology (S.R.V., Y.S.K., H.G.T., J.A.), The Beijer Laboratory, Department of Medicinal Chemistry (J.S.), Department of Medicinal Chemistry (H.A., K.E., U.R.), Science for Life Laboratory, Department of Medicinal Chemistry (M.L.), BMC, Uppsala University, Uppsala, Sweden; Chemical Biology Consortium Sweden, Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medicinal Biochemistry and Biophysics (T.L., A.J.), Karolinska Institute, Sweden; and Biomedicine Discovery Institute, Department of Physiology (L.N., V.P., S.Y.C.), Monash University, Melbourne, Australia
| | - Annika Jenmalm-Jensen
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence (S.D., E.N., A.G., M.H.), Department of Cell and Molecular Biology (S.R.V., Y.S.K., H.G.T., J.A.), The Beijer Laboratory, Department of Medicinal Chemistry (J.S.), Department of Medicinal Chemistry (H.A., K.E., U.R.), Science for Life Laboratory, Department of Medicinal Chemistry (M.L.), BMC, Uppsala University, Uppsala, Sweden; Chemical Biology Consortium Sweden, Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medicinal Biochemistry and Biophysics (T.L., A.J.), Karolinska Institute, Sweden; and Biomedicine Discovery Institute, Department of Physiology (L.N., V.P., S.Y.C.), Monash University, Melbourne, Australia
| | - Hanna Andersson
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence (S.D., E.N., A.G., M.H.), Department of Cell and Molecular Biology (S.R.V., Y.S.K., H.G.T., J.A.), The Beijer Laboratory, Department of Medicinal Chemistry (J.S.), Department of Medicinal Chemistry (H.A., K.E., U.R.), Science for Life Laboratory, Department of Medicinal Chemistry (M.L.), BMC, Uppsala University, Uppsala, Sweden; Chemical Biology Consortium Sweden, Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medicinal Biochemistry and Biophysics (T.L., A.J.), Karolinska Institute, Sweden; and Biomedicine Discovery Institute, Department of Physiology (L.N., V.P., S.Y.C.), Monash University, Melbourne, Australia
| | - Karin Engen
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence (S.D., E.N., A.G., M.H.), Department of Cell and Molecular Biology (S.R.V., Y.S.K., H.G.T., J.A.), The Beijer Laboratory, Department of Medicinal Chemistry (J.S.), Department of Medicinal Chemistry (H.A., K.E., U.R.), Science for Life Laboratory, Department of Medicinal Chemistry (M.L.), BMC, Uppsala University, Uppsala, Sweden; Chemical Biology Consortium Sweden, Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medicinal Biochemistry and Biophysics (T.L., A.J.), Karolinska Institute, Sweden; and Biomedicine Discovery Institute, Department of Physiology (L.N., V.P., S.Y.C.), Monash University, Melbourne, Australia
| | - Ulrika Rosenström
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence (S.D., E.N., A.G., M.H.), Department of Cell and Molecular Biology (S.R.V., Y.S.K., H.G.T., J.A.), The Beijer Laboratory, Department of Medicinal Chemistry (J.S.), Department of Medicinal Chemistry (H.A., K.E., U.R.), Science for Life Laboratory, Department of Medicinal Chemistry (M.L.), BMC, Uppsala University, Uppsala, Sweden; Chemical Biology Consortium Sweden, Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medicinal Biochemistry and Biophysics (T.L., A.J.), Karolinska Institute, Sweden; and Biomedicine Discovery Institute, Department of Physiology (L.N., V.P., S.Y.C.), Monash University, Melbourne, Australia
| | - Mats Larhed
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence (S.D., E.N., A.G., M.H.), Department of Cell and Molecular Biology (S.R.V., Y.S.K., H.G.T., J.A.), The Beijer Laboratory, Department of Medicinal Chemistry (J.S.), Department of Medicinal Chemistry (H.A., K.E., U.R.), Science for Life Laboratory, Department of Medicinal Chemistry (M.L.), BMC, Uppsala University, Uppsala, Sweden; Chemical Biology Consortium Sweden, Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medicinal Biochemistry and Biophysics (T.L., A.J.), Karolinska Institute, Sweden; and Biomedicine Discovery Institute, Department of Physiology (L.N., V.P., S.Y.C.), Monash University, Melbourne, Australia
| | - Johan Åqvist
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence (S.D., E.N., A.G., M.H.), Department of Cell and Molecular Biology (S.R.V., Y.S.K., H.G.T., J.A.), The Beijer Laboratory, Department of Medicinal Chemistry (J.S.), Department of Medicinal Chemistry (H.A., K.E., U.R.), Science for Life Laboratory, Department of Medicinal Chemistry (M.L.), BMC, Uppsala University, Uppsala, Sweden; Chemical Biology Consortium Sweden, Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medicinal Biochemistry and Biophysics (T.L., A.J.), Karolinska Institute, Sweden; and Biomedicine Discovery Institute, Department of Physiology (L.N., V.P., S.Y.C.), Monash University, Melbourne, Australia
| | - Siew Yeen Chai
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence (S.D., E.N., A.G., M.H.), Department of Cell and Molecular Biology (S.R.V., Y.S.K., H.G.T., J.A.), The Beijer Laboratory, Department of Medicinal Chemistry (J.S.), Department of Medicinal Chemistry (H.A., K.E., U.R.), Science for Life Laboratory, Department of Medicinal Chemistry (M.L.), BMC, Uppsala University, Uppsala, Sweden; Chemical Biology Consortium Sweden, Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medicinal Biochemistry and Biophysics (T.L., A.J.), Karolinska Institute, Sweden; and Biomedicine Discovery Institute, Department of Physiology (L.N., V.P., S.Y.C.), Monash University, Melbourne, Australia
| | - Mathias Hallberg
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence (S.D., E.N., A.G., M.H.), Department of Cell and Molecular Biology (S.R.V., Y.S.K., H.G.T., J.A.), The Beijer Laboratory, Department of Medicinal Chemistry (J.S.), Department of Medicinal Chemistry (H.A., K.E., U.R.), Science for Life Laboratory, Department of Medicinal Chemistry (M.L.), BMC, Uppsala University, Uppsala, Sweden; Chemical Biology Consortium Sweden, Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medicinal Biochemistry and Biophysics (T.L., A.J.), Karolinska Institute, Sweden; and Biomedicine Discovery Institute, Department of Physiology (L.N., V.P., S.Y.C.), Monash University, Melbourne, Australia
| |
Collapse
|