1
|
Wang Y, Li M, Haughton D, Kazis LE. Transition of mild cognitive impairment to Alzheimer's disease: Medications as modifiable risk factors. PLoS One 2024; 19:e0306270. [PMID: 39141609 PMCID: PMC11324149 DOI: 10.1371/journal.pone.0306270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 06/13/2024] [Indexed: 08/16/2024] Open
Abstract
BACKGROUND Mild cognitive impairment (MCI) is a pre-clinical stage of Alzheimer's disease (AD). Understanding the transition probabilities across the disease continuum of AD, ranging from MCI to AD to Mortality is crucial for the economic modeling of AD and effective planning of future interventions and healthcare resource allocation decisions. This study uses the Multi-state Markov model to quantify the transition probabilities along the disease progression and specifically investigates medications as modifiable risk factors of AD associated with accelerated or decelerated transition times from MCI to AD, MCI to mortality, and AD to mortality. METHODS Individuals with MCI were identified from the National Alzheimer's Coordinating Center between September 2005 and May 2021. A three-state Markov model was postulated to model the disease progression among three states: MCI, AD, and mortality with adjustment for demographics, genetic characteristics, comorbidities and medications. Transition probabilities, the total length of stay in each state, and the hazard ratios of the use of medications for diabetes, hypertension, and hypercholesterolemia (the known modifiable risk factors of AD) were evaluated for these transitions. RESULTS 3,324 individuals with MCI were identified. The probability of developing AD after one year since the initial diagnosis of MCI is 14.9%. After approximately 6 years from the initial diagnosis of MCI, the probability of transitioning to AD increases to nearly 41.7% before experiencing a subsequent decline. The expected total lengths of stay were 5.38 (95% CI: 0.002-6.03) years at MCI state and 7.61 (95%CI: 0.002-8.88) years at AD state. Patients with active use of lipid-lowering agents were associated with significantly lower hazards of transitioning from MCI to AD (HR: 0.83, 95%CI:0.71-0.96), MCI to mortality (HR: 0.51, 95%CI:0.34-0.77), and AD to mortality (HR: 0.81, 95%CI:0.66-0.99). CONCLUSIONS Results suggest that lipid-lowering agents may confer a protective effect, delaying the onset of AD. Additionally, lipid-lowering agents indicate a favorable association with a longer survival time.
Collapse
Affiliation(s)
- Ying Wang
- Department of Mathematical Sciences, Bentley University, Waltham, Massachusetts, United States of America
- School of Computing and Data Science, Wentworth Institute of Technology, Boston, Massachusetts, United States of America
- Geriatric Research Education and Clinical Center, Bedford VA Healthcare System, Bedford, Massachusetts, United States of America
| | - Mingfei Li
- Department of Mathematical Sciences, Bentley University, Waltham, Massachusetts, United States of America
- Center for Healthcare Organization and Implementation Research, Bedford VA Healthcare System, Bedford, Massachusetts, United States of America
| | - Dominique Haughton
- Department of Mathematical Sciences, Bentley University, Waltham, Massachusetts, United States of America
- Affiliated Researcher, Université Paris 1 (SAMM), Paris, France
- Affiliated Researcher, Université Toulouse 1 (TSE-R), Toulouse, France
| | - Lewis E. Kazis
- Department of Health Law, Policy and Management, Boston University School of Public Health, Boston, Massachusetts, United States of America
- Rehabilitation Outcomes Center (ROC), Spaulding Rehabilitation Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
2
|
Nishida T, Moghimi S, Gunasegaran G, Walker E, Wu JH, Rahmatnejad K, Zangwill LM, Baxter SL, Weinreb RN. Association between metformin use with circumpapillary retinal nerve fibre layer thickness and capillary vessel density in glaucoma. Br J Ophthalmol 2024:bjo-2023-325035. [PMID: 38839252 DOI: 10.1136/bjo-2023-325035] [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: 12/06/2023] [Accepted: 05/21/2024] [Indexed: 06/07/2024]
Abstract
BACKGROUND/AIMS To investigate the association between use of metformin and circumpapillary retinal nerve fibre layer (cpRNFL) thickness, as well as whole image capillary density (wiCD), in patients with glaucoma. METHODS This cross-sectional study included patients with glaucoma suspect or primary open-angle glaucoma (POAG) underwent optical coherence tomography angiography imaging. Use and duration of antidiabetic medications were assessed at the time of imaging. Multivariable linear mixed-effect modelling was used to estimate the effect of diabetes medication on wiCD and cpRNFL while controlling for covariates including age, race, body mass index, diagnosis, 24-2 visual field mean deviation, and intraocular pressure, average signal strength index as well as any variables that showed a p <0.1 in the univariable analysis. RESULTS A total of 577 eyes (330 POAG and 247 glaucoma suspect) of 346 patients were included. Sixty-five patients (23%) had diabetes, of whom 55 (78.5%) used metformin, and 17 (26.2%) used insulin. After adjusting for covariates, the association between metformin use and wiCD (1.56 (95% CI 0.40 to 2.71); p=0.008), duration of metformin use and wiCD (0.12 (95% CI 0.02 to 0.22) per 1 year longer; p=0.037), and metformin use and cpRNFL thickness (5.17 (95% CI 1.24 to 9.10) µm; p=0.010) had statistically significant associations in each model. CONCLUSIONS Metformin use was associated with higher wiCD and thicker cpRNFL. These findings indicate a potential association, underscoring the need for longitudinal studies to determine if metformin plays a role in the retinal conditions of patients with glaucoma. TRIAL REGISTRATION NUMBER NCT00221897.
Collapse
Affiliation(s)
- Takashi Nishida
- Viterbi Family Department of Ophthalmology, University of California San Diego, La Jolla, California, USA
| | - Sasan Moghimi
- Viterbi Family Department of Ophthalmology, University of California San Diego, La Jolla, California, USA
| | - Gopikasree Gunasegaran
- Viterbi Family Department of Ophthalmology, University of California San Diego, La Jolla, California, USA
| | - Evan Walker
- Viterbi Family Department of Ophthalmology, University of California San Diego, La Jolla, California, USA
| | - Jo-Hsuan Wu
- Viterbi Family Department of Ophthalmology, University of California San Diego, La Jolla, California, USA
| | - Kamran Rahmatnejad
- Viterbi Family Department of Ophthalmology, University of California San Diego, La Jolla, California, USA
| | - Linda M Zangwill
- Viterbi Family Department of Ophthalmology, University of California San Diego, La Jolla, California, USA
| | - Sally L Baxter
- Division of Ophthalmology Informatics and Data Science and Hamilton Glaucoma Center, Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California San Diego, La Jolla, California, USA
- Division of Biomedical Informatics, Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Robert N Weinreb
- Viterbi Family Department of Ophthalmology, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
3
|
Zhou Z, Orchard SG, Nelson MR, Fravel MA, Ernst ME. Angiotensin Receptor Blockers and Cognition: a Scoping Review. Curr Hypertens Rep 2024; 26:1-19. [PMID: 37733162 PMCID: PMC10796582 DOI: 10.1007/s11906-023-01266-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/21/2023] [Indexed: 09/22/2023]
Abstract
PURPOSE OF REVIEW To provide an overview of the association between angiotensin II receptor blocker (ARB) use and cognitive outcomes. RECENT FINDINGS ARBs have previously shown greater neuroprotection compared to other anti-hypertensive classes. The benefits are primarily attributed to the ARB's effect on modulating the renin-angiotensin system via inhibiting the Ang II/AT1R pathway and activating the Ang II/AT2R, Ang IV/AT4R, and Ang-(1-7)/MasR pathways. These interactions are associated with pleiotropic neurocognitive benefits, including reduced β-amyloid accumulation and abnormal hyperphosphorylation of tau, ameliorated brain hypo-fusion, reduced neuroinflammation and synaptic dysfunction, better neurotoxin clearing, and blood-brain barrier function restoration. While ACEis also inhibit AT1R, they simultaneously lower Ang II and block the Ang II/AT2R and Ang IV/AT4R pathways that counterbalance the potential benefits. ARBs may be considered an adjunctive approach for neuroprotection. This preliminary evidence, coupled with their underlying mechanistic pathways, emphasizes the need for future long-term randomized trials to yield more definitive results.
Collapse
Affiliation(s)
- Zhen Zhou
- School of Public Health and Preventive Medicine, Monash University, 553 St Kilda Road, Melbourne, VIC, 3004, Australia.
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia.
| | - Suzanne G Orchard
- School of Public Health and Preventive Medicine, Monash University, 553 St Kilda Road, Melbourne, VIC, 3004, Australia
| | - Mark R Nelson
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Michelle A Fravel
- Department of Pharmacy Practice and Science, College of Pharmacy, The University of Iowa, Iowa, IA, USA
| | - Michael E Ernst
- Department of Pharmacy Practice and Science, College of Pharmacy, The University of Iowa, Iowa, IA, USA.
- Department of Family Medicine, Carver College of Medicine, 01291-A PFP, The University of Iowa, 200 Hawkins Dr, Iowa, IA, 52242, USA.
| |
Collapse
|
4
|
Fleisher-Berkovich S, Battaglia V, Baratta F, Brusa P, Ventura Y, Sharon N, Dahan A, Collino M, Ben-Shabat S. An Emerging Strategy for Neuroinflammation Treatment: Combined Cannabidiol and Angiotensin Receptor Blockers Treatments Effectively Inhibit Glial Nitric Oxide Release. Int J Mol Sci 2023; 24:16254. [PMID: 38003444 PMCID: PMC10671332 DOI: 10.3390/ijms242216254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/06/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
Cannabidiol (CBD), the major non-psychoactive phytocannabinoid found in cannabis, has anti-neuroinflammatory properties. Despite the increasing use of CBD, little is known about its effect in combination with other substances. Combination therapy has been gaining attention recently, aiming to produce more efficient effects. Angiotensin II activates the angiotensin 1 receptor and regulates neuroinflammation and cognition. Angiotensin receptor 1 blockers (ARBs) were shown to be neuroprotective and prevent cognitive decline. The present study aimed to elucidate the combined role of CBD and ARBs in the modulation of lipopolysaccharide (LPS)-induced glial inflammation. While LPS significantly enhanced nitric oxide synthesis vs. the control, telmisartan and CBD, when administered alone, attenuated this effect by 60% and 36%, respectively. Exposure of LPS-stimulated cells to both compounds resulted in the 95% inhibition of glial nitric oxide release (additive effect). A synergistic inhibitory effect on nitric oxide release was observed when cells were co-treated with losartan (5 μM) and CBD (5 μM) (by 80%) compared to exposure to each compound alone (by 22% and 26%, respectively). Telmisartan and CBD given alone increased TNFα levels by 60% and 40%, respectively. CBD and telmisartan, when given together, attenuated the LPS-induced increase in TNFα levels without statistical significance. LPS-induced IL-17 release was attenuated by CBD with or without telmisartan (by 75%) or telmisartan alone (by 60%). LPS-induced Interferon-γ release was attenuated by 80% when telmisartan was administered in the absence or presence of CBD. Anti-inflammatory effects were recorded when CBD was combined with the known anti-inflammatory agent dimethyl fumarate (DMF)/monomethyl fumarate (MMF). A synergistic inhibitory effect of CBD and MMF on glial release of nitric oxide (by 77%) was observed compared to cells exposed to MMF (by 35%) or CBD (by 12%) alone. Overall, this study highlights the potential of new combinations of CBD (5 μM) with losartan (5 μM) or MMF (1 μM) to synergistically attenuate glial NO synthesis. Additive effects on NO production were observed when telmisartan (5 μM) and CBD (5 μM) were administered together to glial cells.
Collapse
Affiliation(s)
- Sigal Fleisher-Berkovich
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 8410501, Israel; (Y.V.); (N.S.); (A.D.)
| | - Veronica Battaglia
- Department of Drug Science and Technology, University of Torino, 10124 Torino, Italy; (V.B.); (F.B.); (P.B.)
| | - Francesca Baratta
- Department of Drug Science and Technology, University of Torino, 10124 Torino, Italy; (V.B.); (F.B.); (P.B.)
| | - Paola Brusa
- Department of Drug Science and Technology, University of Torino, 10124 Torino, Italy; (V.B.); (F.B.); (P.B.)
| | - Yvonne Ventura
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 8410501, Israel; (Y.V.); (N.S.); (A.D.)
| | - Nitzan Sharon
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 8410501, Israel; (Y.V.); (N.S.); (A.D.)
| | - Arik Dahan
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 8410501, Israel; (Y.V.); (N.S.); (A.D.)
| | - Massimo Collino
- Department of Neuroscience “Rita Levi Montalcini”, University of Torino, 10124 Torino, Italy;
| | - Shimon Ben-Shabat
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 8410501, Israel; (Y.V.); (N.S.); (A.D.)
| |
Collapse
|
5
|
Murase S, Sakitani N, Maekawa T, Yoshino D, Takano K, Konno A, Hirai H, Saito T, Tanaka S, Shinohara K, Kishi T, Yoshikawa Y, Sakai T, Ayaori M, Inanami H, Tomiyasu K, Takashima A, Ogata T, Tsuchimochi H, Sato S, Saito S, Yoshino K, Matsuura Y, Funamoto K, Ochi H, Shinohara M, Nagao M, Sawada Y. Interstitial-fluid shear stresses induced by vertically oscillating head motion lower blood pressure in hypertensive rats and humans. Nat Biomed Eng 2023; 7:1350-1373. [PMID: 37414976 PMCID: PMC10651490 DOI: 10.1038/s41551-023-01061-x] [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: 06/10/2020] [Accepted: 05/27/2023] [Indexed: 07/08/2023]
Abstract
The mechanisms by which physical exercise benefits brain functions are not fully understood. Here, we show that vertically oscillating head motions mimicking mechanical accelerations experienced during fast walking, light jogging or treadmill running at a moderate velocity reduce the blood pressure of rats and human adults with hypertension. In hypertensive rats, shear stresses of less than 1 Pa resulting from interstitial-fluid flow induced by such passive head motions reduced the expression of the angiotensin II type-1 receptor in astrocytes in the rostral ventrolateral medulla, and the resulting antihypertensive effects were abrogated by hydrogel introduction that inhibited interstitial-fluid movement in the medulla. Our findings suggest that oscillatory mechanical interventions could be used to elicit antihypertensive effects.
Collapse
Affiliation(s)
- Shuhei Murase
- Department of Rehabilitation for Motor Functions, National Rehabilitation Center for Persons with Disabilities, Tokorozawa, Japan
- Department of Orthopaedic Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Naoyoshi Sakitani
- Department of Rehabilitation for Motor Functions, National Rehabilitation Center for Persons with Disabilities, Tokorozawa, Japan
- Department of Cell Biology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Takahiro Maekawa
- Department of Rehabilitation for Motor Functions, National Rehabilitation Center for Persons with Disabilities, Tokorozawa, Japan
| | - Daisuke Yoshino
- Division of Advanced Applied Physics, Institute of Engineering, Tokyo University of Agriculture and Technology, Koganei, Japan
| | - Kouji Takano
- Department of Rehabilitation for Brain Functions, National Rehabilitation Center for Persons with Disabilities, Tokorozawa, Japan
| | - Ayumu Konno
- Department of Neurophysiology & Neural Repair, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Hirokazu Hirai
- Department of Neurophysiology & Neural Repair, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Taku Saito
- Department of Orthopaedic Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Sakae Tanaka
- Department of Orthopaedic Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Keisuke Shinohara
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takuya Kishi
- Department of Cardiology, Graduate School of Medicine, International University of Health and Welfare, Okawa, Japan
| | - Yuki Yoshikawa
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Takamasa Sakai
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | | | - Hirohiko Inanami
- Inanami Spine & Joint Hospital/Iwai Orthopaedic Medical Hospital, Iwai Medical Foundation, Tokyo, Japan
| | - Koji Tomiyasu
- Center of Sports Science and Health Promotion, National Rehabilitation Center for Persons with Disabilities, Tokorozawa, Japan
| | - Atsushi Takashima
- Department of Assistive Technology, National Rehabilitation Center for Persons with Disabilities, Tokorozawa, Japan
| | - Toru Ogata
- Department of Rehabilitation for Motor Functions, National Rehabilitation Center for Persons with Disabilities, Tokorozawa, Japan
- Department of Rehabilitation Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hirotsugu Tsuchimochi
- Department of Cardiac Physiology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Shinya Sato
- Department of Advanced Medical Technologies, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Shigeyoshi Saito
- Department of Medical Physics and Engineering, Division of Health Sciences, Osaka University Graduate School of Medicine, Suita, Japan
| | - Kohzoh Yoshino
- School of Biological and Environmental Sciences, Kwansei Gakuin University, Sanda, Japan
| | - Yuiko Matsuura
- Department of Health and Sports, Niigata University of Health and Welfare, Niigata, Japan
| | | | - Hiroki Ochi
- Department of Rehabilitation for Motor Functions, National Rehabilitation Center for Persons with Disabilities, Tokorozawa, Japan
| | - Masahiro Shinohara
- Department of Rehabilitation for Motor Functions, National Rehabilitation Center for Persons with Disabilities, Tokorozawa, Japan
| | - Motoshi Nagao
- Department of Rehabilitation for Motor Functions, National Rehabilitation Center for Persons with Disabilities, Tokorozawa, Japan
| | - Yasuhiro Sawada
- Department of Rehabilitation for Motor Functions, National Rehabilitation Center for Persons with Disabilities, Tokorozawa, Japan.
- Department of Orthopaedic Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
- Department of Cell Biology, National Cerebral and Cardiovascular Center, Suita, Japan.
- Division of Advanced Applied Physics, Institute of Engineering, Tokyo University of Agriculture and Technology, Koganei, Japan.
- Department of Clinical Research, National Rehabilitation Center for Persons with Disabilities, Tokorozawa, Japan.
| |
Collapse
|
6
|
Villapol S, Janatpour ZC, Affram KO, Symes AJ. The Renin Angiotensin System as a Therapeutic Target in Traumatic Brain Injury. Neurotherapeutics 2023; 20:1565-1591. [PMID: 37759139 PMCID: PMC10684482 DOI: 10.1007/s13311-023-01435-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2023] [Indexed: 09/29/2023] Open
Abstract
Traumatic brain injury (TBI) is a major public health problem, with limited pharmacological options available beyond symptomatic relief. The renin angiotensin system (RAS) is primarily known as a systemic endocrine regulatory system, with major roles controlling blood pressure and fluid homeostasis. Drugs that target the RAS are used to treat hypertension, heart failure and kidney disorders. They have now been used chronically by millions of people and have a favorable safety profile. In addition to the systemic RAS, it is now appreciated that many different organ systems, including the brain, have their own local RAS. The major ligand of the classic RAS, Angiotensin II (Ang II) acts predominantly through the Ang II Type 1 receptor (AT1R), leading to vasoconstriction, inflammation, and heightened oxidative stress. These processes can exacerbate brain injuries. Ang II receptor blockers (ARBs) are AT1R antagonists. They have been shown in several preclinical studies to enhance recovery from TBI in rodents through improvements in molecular, cellular and behavioral correlates of injury. ARBs are now under consideration for clinical trials in TBI. Several different RAS peptides that signal through receptors distinct from the AT1R, are also potential therapeutic targets for TBI. The counter regulatory RAS pathway has actions that oppose those stimulated by AT1R signaling. This alternative pathway has many beneficial effects on cells in the central nervous system, bringing about vasodilation, and having anti-inflammatory and anti-oxidative stress actions. Stimulation of this pathway also has potential therapeutic value for the treatment of TBI. This comprehensive review will provide an overview of the various components of the RAS, with a focus on their direct relevance to TBI pathology. It will explore different therapeutic agents that modulate this system and assess their potential efficacy in treating TBI patients.
Collapse
Affiliation(s)
- Sonia Villapol
- Department of Neurosurgery, Houston Methodist Hospital, Houston, TX, USA
| | - Zachary C Janatpour
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Kwame O Affram
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
| | - Aviva J Symes
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
| |
Collapse
|
7
|
Scheinman SB, Tseng KY, Alford S, Tai LM. Higher Neuronal Facilitation and Potentiation with APOE4 Suppressed by Angiotensin II. RESEARCH SQUARE 2023:rs.3.rs-2960437. [PMID: 37292788 PMCID: PMC10246245 DOI: 10.21203/rs.3.rs-2960437/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Progressive hippocampal degeneration is a key component of Alzheimer's disease (AD) progression. Therefore, identifying how hippocampal neuronal function is modulated early in AD is an important approach to eventually prevent degeneration. AD-risk factors and signaling molecules likely modulate neuronal function, including APOE genotype and angiotensin II. Compared to APOE3 , APOE4 increases AD risk up to 12-fold, and high levels of angiotensin II are hypothesized to disrupt neuronal function in AD. However, the extent that APOE and angiotensin II modulates the hippocampal neuronal phenotype in AD-relevant models is unknown. To address this issue, we used electrophysiological techniques to assess the impact of APOE genotype and angiotensin II on basal synaptic transmission, presynaptic and post-synaptic activity in mice that express human APOE3 (E3FAD) or APOE4 (E4FAD) and overproduce Aβ. We found that compared to E3FAD mice, E4FAD mice had lower basal synaptic activity, but higher levels of paired pulse facilitation (PPF) and Long-Term Potentiation (LTP) in the Schaffer Collateral Commissural Pathway (SCCP) of the hippocampus. We also found that exogenous angiotensin II has a profound inhibitory effect on hippocampal LTP in both E3FAD and E4FAD mice. Collectively, our data suggests that APOE4 and Aβ are associated with a hippocampal phenotype comprised of lower basal activity and higher responses to high frequency stimulation, the latter of which is suppressed by angiotensin II. These novel data suggest a potential mechanistic link between hippocampal activity, APOE4 genotype and angiotensin II in AD.
Collapse
Affiliation(s)
| | - Kuei Y Tseng
- University of Illinois at Chicago College of Medicine
| | - Simon Alford
- University of Illinois at Chicago College of Medicine
| | - Leon M Tai
- University of Illinois at Chicago College of Medicine
| |
Collapse
|
8
|
Pan X, Yun J, Coban Akdemir ZH, Jiang X, Wu E, Huang JH, Sahni N, Yi SS. AI-DrugNet: A network-based deep learning model for drug repurposing and combination therapy in neurological disorders. Comput Struct Biotechnol J 2023; 21:1533-1542. [PMID: 36879885 PMCID: PMC9984442 DOI: 10.1016/j.csbj.2023.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 02/03/2023] [Accepted: 02/03/2023] [Indexed: 02/10/2023] Open
Abstract
Discovering effective therapies is difficult for neurological and developmental disorders in that disease progression is often associated with a complex and interactive mechanism. Over the past few decades, few drugs have been identified for treating Alzheimer's disease (AD), especially for impacting the causes of cell death in AD. Although drug repurposing is gaining more success in developing therapeutic efficacy for complex diseases such as common cancer, the complications behind AD require further study. Here, we developed a novel prediction framework based on deep learning to identify potential repurposed drug therapies for AD, and more importantly, our framework is broadly applicable and may generalize to identifying potential drug combinations in other diseases. Our prediction framework is as follows: we first built a drug-target pair (DTP) network based on multiple drug features and target features, as well as the associations between DTP nodes where drug-target pairs are the DTP nodes and the associations between DTP nodes are represented as the edges in the AD disease network; furthermore, we incorporated the drug-target feature from the DTP network and the relationship information between drug-drug, target-target, drug-target within and outside of drug-target pairs, representing each drug-combination as a quartet to generate corresponding integrated features; finally, we developed an AI-based Drug discovery Network (AI-DrugNet), which exhibits robust predictive performance. The implementation of our network model help identify potential repurposed and combination drug options that may serve to treat AD and other diseases.
Collapse
Affiliation(s)
- Xingxin Pan
- Livestrong Cancer Institutes, Department of Oncology, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA
| | - Jun Yun
- Oden Institute for Computational Engineering and Sciences (ICES), The University of Texas at Austin, Austin, TX 78712, USA
| | - Zeynep H. Coban Akdemir
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Xiaoqian Jiang
- School of Biomedical Informatics, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Erxi Wu
- Livestrong Cancer Institutes, Department of Oncology, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA
- Neuroscience Institute and Department of Neurosurgery, Baylor Scott & White Health, Temple, TX 76502, USA
- Department of Surgery, Texas A & M University Health Science Center, College of Medicine, Temple, TX 76508, USA
- Department of Pharmaceutical Sciences, Texas A & M University Health Science Center, College of Pharmacy, College Station, TX 77843, USA
| | - Jason H. Huang
- Neuroscience Institute and Department of Neurosurgery, Baylor Scott & White Health, Temple, TX 76502, USA
- Department of Surgery, Texas A & M University Health Science Center, College of Medicine, Temple, TX 76508, USA
| | - Nidhi Sahni
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Smithville, TX 78957, USA
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Quantitative and Computational Biosciences Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - S. Stephen Yi
- Livestrong Cancer Institutes, Department of Oncology, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA
- Oden Institute for Computational Engineering and Sciences (ICES), The University of Texas at Austin, Austin, TX 78712, USA
- Interdisciplinary Life Sciences Graduate Programs (ILSGP), College of Natural Sciences, The University of Texas at Austin, Austin, TX 78712, USA
- Department of Biomedical Engineering, Cockrell School of Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
9
|
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
|
10
|
Chrysant SG. Superior stroke prevention with angiotensin receptor blockers compared with other antihypertensive drugs. Expert Opin Drug Saf 2023; 22:125-131. [PMID: 36882886 DOI: 10.1080/14740338.2023.2189236] [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: 03/09/2023]
Abstract
INTRODUCTION Stroke is a major cause of death and disability and its incidence is linearly increased with the elevation of blood pressure (BP) and the advancement of age in both men and women, with its incidence being higher in older subjects, the blacks and women. AREAS COVERED The annual worldwide incidence of stroke is 7.6 million for subjects ≥ 20 years of age with the average direct and indirect annual costs of stroke care, is expected to be $94.3 billion between 2014 and 2015. With respect to the cause of stroke, this is multifactorial, due to atherosclerotic heart disease, inflammation, atrial fibrillation, and hypertension with the latter being the most important cause. Therefore, control of BP is the major factor for its prevention. In order to get a better perspective on the current management of stroke, a Medline search of the English literature was conducted between 2014 and 2022 and 26 pertinent papers were selected. EXPERT OPINION Review of data from the selected papers demonstrated that control of SSBP < 130 mmHg was better in stroke prevention than SBP 130-140 mmHg for primary and secondary strokes. Among the drugs used, angiotensin receptor blockers provided superior stroke prevention compared to angiotensin converting enzyme inhibitors and other antihypertensive drugs.
Collapse
Affiliation(s)
- Steven G Chrysant
- Department of Cardiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.,Department of Cardiology, INTEGRIS Baptist Medical Center, Oklahoma City, OK, USA
| |
Collapse
|
11
|
D’Silva E, Meor Azlan NF, Zhang J. Angiotensin II Receptor Blockers in the Management of Hypertension in Preventing Cognitive Impairment and Dementia-A Systematic Review. Pharmaceutics 2022; 14:pharmaceutics14102123. [PMID: 36297558 PMCID: PMC9609033 DOI: 10.3390/pharmaceutics14102123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/01/2022] [Accepted: 10/03/2022] [Indexed: 11/18/2022] Open
Abstract
Hypertension is a known risk factor for cognition-related pathologies including dementia. The National Institute of Health and Care Excellence (NICE) guidelines recommend angiotensin (Ang) II receptor blockers (ARBs) or angiotensin-converting enzyme inhibitors (ACEIs) as a first-line treatment for hypertension. Although both ARBs and ACEIs show neuroprotective effects, ACEIs show contradictory side effects; therefore, ARBs may be a more viable option. However, trials assessing the effects of ARBs on cognition are scarce and conflicting. Therefore, the aim of this review is to conduct a systematic review and synthesise data on the influence of ARBs on cognition and dementia prevention. Five databases were searched from 1992-2022 to produce 13 randomised controlled trials (RCTs) involving 26,907 patients that compared associations of ARBs against placebos or other antihypertensives on cognition or probable dementia with a minimum duration of 3 months. ARBs showed greater cognitive benefits when compared to hydrochlorothiazide (HCTZ), beta blockers (BB), and ACEIs. Our findings showed that although ARBs are superior to some antihypertensives such as ACEIs, thiazide and beta blockers, they made no difference in comparison to the placebo in all but one sample of patients. The positive effects on cognitive performances are equal to calcium channel blockers (CCBs) and lower than statin. The neuroprotective effects of ARBs are also more beneficial when ARBs are taken at the same time as a statin. Due to these inconsistencies, robust conclusions cannot be made. Future trials are warranted and, if successful, could have positive economic implications and consequently improve quality of life.
Collapse
Affiliation(s)
- Elvira D’Silva
- Hatherly Laboratories, Institute of Biomedical and Clinical Sciences, Medical School, Faculty of Health and Life Sciences, Streatham Campus, University of Exeter, Exeter EX4 4PS, UK
| | - Nur Farah Meor Azlan
- Hatherly Laboratories, Institute of Biomedical and Clinical Sciences, Medical School, Faculty of Health and Life Sciences, Streatham Campus, University of Exeter, Exeter EX4 4PS, UK
| | - Jinwei Zhang
- Hatherly Laboratories, Institute of Biomedical and Clinical Sciences, Medical School, Faculty of Health and Life Sciences, Streatham Campus, University of Exeter, Exeter EX4 4PS, UK
- School of Medicine, Xiamen Cardiovascular Hospital of Xiamen University, Xiamen University, Xiang’an Nan Lu, Xiamen 361102, China
- State Key Laboratory of Bioorganic & Natural Products Chemistry, Research Center of Chemical Kinomics, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, # 345 Lingling Road, Shanghai 200032, China
- Correspondence: ; Tel.: +44-0-1392-72-3828
| |
Collapse
|
12
|
Russo M, De Rosa MA, Calisi D, Consoli S, Evangelista G, Dono F, Santilli M, Granzotto A, Onofrj M, Sensi SL. Migraine Pharmacological Treatment and Cognitive Impairment: Risks and Benefits. Int J Mol Sci 2022; 23:11418. [PMID: 36232720 PMCID: PMC9569564 DOI: 10.3390/ijms231911418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 11/23/2022] Open
Abstract
Migraine is a common neurological disorder impairing the quality of life of patients. The condition requires, as an acute or prophylactic line of intervention, the frequent use of drugs acting on the central nervous system (CNS). The long-term impact of these medications on cognition and neurodegeneration has never been consistently assessed. The paper reviews pharmacological migraine treatments and discusses their biological and clinical effects on the CNS. The different anti-migraine drugs show distinct profiles concerning neurodegeneration and the risk of cognitive deficits. These features should be carefully evaluated when prescribing a pharmacological treatment as many migraineurs are of scholar or working age and their performances may be affected by drug misuse. Thus, a reconsideration of therapy guidelines is warranted. Furthermore, since conflicting results have emerged in the relationship between migraine and dementia, future studies must consider present and past pharmacological regimens as potential confounding factors.
Collapse
Affiliation(s)
- Mirella Russo
- Department of Neurosciences, Imaging and Clinical Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- CAST—Center for Advanced Studies and Technology, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Matteo A. De Rosa
- Department of Neurosciences, Imaging and Clinical Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Dario Calisi
- Department of Neurosciences, Imaging and Clinical Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Stefano Consoli
- Department of Neurosciences, Imaging and Clinical Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Giacomo Evangelista
- Department of Neurosciences, Imaging and Clinical Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Fedele Dono
- Department of Neurosciences, Imaging and Clinical Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- CAST—Center for Advanced Studies and Technology, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Matteo Santilli
- Department of Neurosciences, Imaging and Clinical Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Alberto Granzotto
- Department of Neurosciences, Imaging and Clinical Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- CAST—Center for Advanced Studies and Technology, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Marco Onofrj
- Department of Neurosciences, Imaging and Clinical Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- CAST—Center for Advanced Studies and Technology, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Stefano L. Sensi
- Department of Neurosciences, Imaging and Clinical Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- CAST—Center for Advanced Studies and Technology, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- Institute for Mind Impairments and Neurological Disorders-iMIND, University of California, Irvine, Irvine, CA 92697, USA
- ITAB—Institute of Advanced Biomedical Technology, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| |
Collapse
|
13
|
Liampas I, Hatzimanolis A, Siokas V, Yannakoulia M, Kosmidis MH, Sakka P, Hadjigeorgiou GM, Scarmeas N, Dardiotis E. Antihypertensive Medication Class and the Risk of Dementia and Cognitive Decline in Older Adults: A Secondary Analysis of the Prospective HELIAD Cohort. J Alzheimers Dis 2022; 89:709-719. [PMID: 35912747 DOI: 10.3233/jad-220439] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND It is unclear whether the main antihypertensive medication classes (diuretics, calcium channel blockers, beta-blockers, angiotensin converting enzyme inhibitors, and angiotensin receptor blockers (ARBs)) are associated with different risks of cognitive decline. Published evidence is conflicting and stems mainly from observational studies. OBJECTIVE To investigate the differential effects of antihypertensives on the risks of developing dementia and cognitive decline, with a specific focus on the vascular component of the mechanisms underlying these interactions. METHODS Older adults with a history of hypertension and without dementia were drawn from the population-based HELIAD cohort. Age-, gender-, education-, and antihypertensive medication- (five dichotomous exposures) adjusted Cox proportional-hazards models and generalized estimating equations were performed to appraise the associations of baseline antihypertensive therapy with dementia incidence and cognitive decline (quantified using a comprehensive neuropsychological battery). Analyses were subsequently adjusted for clinical vascular risk (dyslipidemia, diabetes mellitus, smoking, cardiovascular, and cerebrovascular history) and genetic susceptibility to stroke (using polygenic risk scores generated according to the MEGASTROKE consortium GWAS findings). RESULTS A total of 776 predominantly female participants (73.61±4.94 years) with hypertension and a mean follow-up of 3.02±0.82 years were analyzed. Baseline treatment was not associated with the risk of incident dementia. ARB users experienced a slower yearly global cognitive [2.5% of a SD, 95% CI = (0.1, 4.9)] and language [4.4% of a SD, 95% CI = (1.4, 7.4)] decline compared to non-users. The fully adjusted model reproduced similar associations for both global cognitive [β= 0.027, 95% CI = (-0.003, 0.057)], and language decline [β= 0.063, 95% CI = (0.023, 0.104)]. CONCLUSION ARBs may be superior to other antihypertensive agents in the preservation of cognition, an association probably mediated by vascular-independent mechanisms.
Collapse
Affiliation(s)
- Ioannis Liampas
- Department of Neurology, University Hospital of Larissa, School of Medicine, University of Thessaly, Larissa, Greece
| | - Alex Hatzimanolis
- Department of Psychiatry, Aiginition Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Vasileios Siokas
- Department of Neurology, University Hospital of Larissa, School of Medicine, University of Thessaly, Larissa, Greece
| | - Mary Yannakoulia
- Department of Nutrition and Dietetics, Harokopio University, Athens, Greece
| | - Mary H Kosmidis
- Lab of Cognitive Neuroscience, School of Psychology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Paraskevi Sakka
- Athens Association of Alzheimer's Disease and Related Disorders, Marousi, Athens, Greece
| | - Georgios M Hadjigeorgiou
- Department of Neurology, University Hospital of Larissa, School of Medicine, University of Thessaly, Larissa, Greece.,Department of Neurology, Medical School, University of Cyprus, Nicosia, Cyprus
| | - Nikolaos Scarmeas
- 1st Department of Neurology, Aiginition Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece.,Taub Institute for Research in Alzheimer's Disease and the Aging Brain, The Gertrude H. Sergievsky Center, Department of Neurology, Columbia University, New York, NY, USA
| | - Efthimios Dardiotis
- Department of Neurology, University Hospital of Larissa, School of Medicine, University of Thessaly, Larissa, Greece
| |
Collapse
|
14
|
Sheng J, Zu Z, Zhang Y, Zhu H, Qi J, Zheng T, Tian Y, Zhang L. Targeted therapy of atherosclerosis by zeolitic imidazolate framework-8 nanoparticles loaded with losartan potassium via simultaneous lipid-scavenging and anti-inflammation. J Mater Chem B 2022; 10:5925-5937. [PMID: 35639392 DOI: 10.1039/d2tb00686c] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Atherosclerosis (AS) is a condition associated with dysfunctional lipid metabolism and an inflammatory immune microenvironment that remains the leading cause of severe cardiovascular events. Drugs exhibiting both anti-inflammatory and lipid-scavenging...
Collapse
Affiliation(s)
- Jie Sheng
- Department of Radiology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Ziyue Zu
- Department of Radiology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Yugang Zhang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Centre of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Haitao Zhu
- Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, 212001, China
| | - Jianchen Qi
- Department of Radiology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Tao Zheng
- Department of Radiology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Ying Tian
- Department of Radiology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Longjiang Zhang
- Department of Radiology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| |
Collapse
|
15
|
Chakraborty A, Sami SA, Marma KKS. A comprehensive review on RAGE-facilitated pathological pathways connecting Alzheimer’s disease, diabetes mellitus, and cardiovascular diseases. THE EGYPTIAN JOURNAL OF INTERNAL MEDICINE 2021. [DOI: 10.1186/s43162-021-00081-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Alzheimer’s disease (AD), cardiovascular disease (CVD), and diabetes are some of the most common causes of morbidity and mortality among the aging populations and cause a heavy burden on the worldwide healthcare system. In this review, we briefly highlighted cellular inflammation-based pathways of diabetes mellitus and CVD through receptor for advanced glycation end products AGEs or RAGE leading to Alzheimer’s disease and interrelation between these vascular and metabolic disorders. The articles were retrieved from Google Scholar, Science Direct, and PubMed databases using the following terms: Alzheimer’s; AGEs; RAGE; RAGE in Alzheimer’s; AGEs in Alzheimer’s; RAGE in diabetes; RAGE related pathways of CVD; RAGE in hypertension; RAGE and RAS system; RAGE and oxidative stress.
Main body of the abstract
AD is a neurodegenerative disease characterized by cognitive dysfunction and neuronal cell death. Vascular complications like hypertension, coronary artery disease, and atherosclerosis as well as metabolic syndromes like obesity and diabetes are related to the pathophysiology of AD. RAGE plays significant role in the onset and progression of AD. Amyloid plaques and neurofibrillary tangles (NFT) are two main markers of AD that regulates via RAGE and other RAGE/ligands interactions which also induces oxidative stress and a cascade of other cellular inflammation pathways leading to AD. Though AD and diabetes are two different disorders but may be inter-linked by AGEs and RAGE. In long-term hyperglycemia, upregulated AGEs interacts with RAGE and produces reactive oxygen species which induces further inflammation and vascular complications. Aging, hypercholesterolemia, atherosclerosis, hypertension, obesity, and inflammation are some of the main risk factors for both diabetes and dementia. Chronic hypertension and coronary artery disease disrupt the functions of the blood-brain barrier and are responsible for the accumulation of senile plaques and NFTs.
Short conclusion
RAGE plays a role in the etiology of Aβ and tau hyperphosphorylation, both of which contribute to cognitive impairment. So far, targeting RAGE may provide a potential sight to develop therapies against some metabolic disorders.
Collapse
|
16
|
Chen W, Jiang L, Hu Y, Fang G, Yang B, Li J, Liang N, Wu L, Hussain Z. Nanomedicines, an emerging therapeutic regimen for treatment of ischemic cerebral stroke: A review. J Control Release 2021; 340:342-360. [PMID: 34695522 DOI: 10.1016/j.jconrel.2021.10.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 10/14/2021] [Accepted: 10/15/2021] [Indexed: 12/18/2022]
Abstract
Owing to its intricate pathophysiology, cerebral stroke is a serious medical condition caused by interruption or obstruction of blood supply (blockage of vasculature) to the brain tissues which results in diminished supply of essential nutrients and oxygen (hypoxia) and ultimate necrosis of neuronal tissues. A prompt risks assessment and immediate rational therapeutic plan with proficient neuroprotection play critically important role in the effective management of this neuronal emergency. Various conventional medications are being used for treatment of acute ischemic cerebral stroke but fibrinolytic agents, alone or in combination with other agents are considered the mainstay. These clot-busting agents effectively restore blood supply (reperfusion) to ischemic regions of the brain; however, their clinical significance is hampered due to various factors such as short plasma half-life, limited distribution to brain tissues due to the presence of highly efficient physiological barrier, blood brain barrier (BBB), and lacking of target-specific delivery to the ischemic brain regions. To alleviate these issues, various types of nanomedicines such as polymeric nanoparticles (NPs), liposomes, nanoemulsion, micelles and dendrimers have been designed and evaluated. The implication of these newer therapies (nanomedicines) have revolutionized the therapeutic outcomes by improving the plasma half-life, permeation across BBB, efficient distribution to ischemic cerebral tissues and neuroprotection. Furthermore, the adaptation of some diverse techniques including PEGylation, tethering of targeting ligands on the surfaces of nanomedicines, and pH responsive features have also been pondered. The implication of these emerging adaptations have shown remarkable potential in maximizing the targeting efficiency of drugs to ischemic brain tissues, simultaneous delivery of drugs and imaging agents (for early prognosis as well as monitoring of therapy), and therapeutic outcomes such as long-term neuroprotection.
Collapse
Affiliation(s)
- Wei Chen
- Department of Neurology, The First Affiliated Hospital of Guangxi, University of Chinese Medicine, Nanning, Guangxi 530023, China; Graduate School, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, China
| | - Lingfei Jiang
- Graduate College, Guangxi University of Chinese Medicine, Nanning, Guangxi 530200, China
| | - Yueqiang Hu
- Department of Neurology, The First Affiliated Hospital of Guangxi, University of Chinese Medicine, Nanning, Guangxi 530023, China; Guangxi Key Laboratory of Chinese Medicine Foundation Research, Guangxi University of Chinese Medicine, Nanning, Guangxi 530200, China.
| | - Gang Fang
- Guangxi Zhuang and Yao Medicine Engineering Technology Research Center, Guangxi University of Chinese Medicine, Nanning, Guangxi 530200, China
| | - Bilin Yang
- Graduate College, Guangxi University of Chinese Medicine, Nanning, Guangxi 530200, China
| | - Junhong Li
- Department of Neurology, The First Affiliated Hospital of Guangxi, University of Chinese Medicine, Nanning, Guangxi 530023, China
| | - Ni Liang
- Department of Neurology, The First Affiliated Hospital of Guangxi, University of Chinese Medicine, Nanning, Guangxi 530023, China
| | - Lin Wu
- Department of Neurology, The First Affiliated Hospital of Guangxi, University of Chinese Medicine, Nanning, Guangxi 530023, China; Guangxi Key Laboratory of Chinese Medicine Foundation Research, Guangxi University of Chinese Medicine, Nanning, Guangxi 530200, China.
| | - Zahid Hussain
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates; Research Institute for Medical & Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates.
| |
Collapse
|
17
|
Boily M, Li L, Vallerand D, Girouard H. Angiotensin II Disrupts Neurovascular Coupling by Potentiating Calcium Increases in Astrocytic Endfeet. J Am Heart Assoc 2021; 10:e020608. [PMID: 34459216 PMCID: PMC8649296 DOI: 10.1161/jaha.120.020608] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 07/09/2021] [Indexed: 11/16/2022]
Abstract
Background Angiotensin II (Ang II), a critical mediator of hypertension, impairs neurovascular coupling. Since astrocytes are key regulators of neurovascular coupling, we sought to investigate whether Ang II impairs neurovascular coupling through modulation of astrocytic Ca2+ signaling. Methods and Results Using laser Doppler flowmetry, we found that Ang II attenuates cerebral blood flow elevations induced by whisker stimulation or the metabotropic glutamate receptors agonist, 1S, 3R-1-aminocyclopentane-trans-1,3-dicarboxylic acid (P<0.01). In acute brain slices, Ang II shifted the vascular response induced by 1S, 3R-1-aminocyclopentane-trans-1,3-dicarboxylic acid towards vasoconstriction (P<0.05). The resting and 1S, 3R-1-aminocyclopentane-trans-1,3-dicarboxylic acid-induced Ca2+ levels in the astrocytic endfeet were more elevated in the presence of Ang II (P<0.01). Both effects were reversed by the AT1 receptor antagonist, candesartan (P<0.01 for diameter and P<0.05 for calcium levels). Using photolysis of caged Ca2+ in astrocytic endfeet or pre-incubation of 1,2-Bis(2-aminophenoxy)ethane-N,N,N',N'-tetra-acetic acid tetrakis (acetoxymethyl ester), we demonstrated the link between potentiated Ca2+ elevation and impaired vascular response in the presence of Ang II (P<0.001 and P<0.05, respectively). Both intracellular Ca2+ mobilization and Ca2+ influx through transient receptor potential vanilloid 4 mediated Ang II-induced astrocytic Ca2+ elevation, since blockade of these pathways significantly prevented the intracellular Ca2+ in response to 1S, 3R-1-aminocyclopentane-trans-1,3-dicarboxylic acid (P<0.05). Conclusions These results suggest that Ang II through its AT1 receptor potentiates the astrocytic Ca2+ responses to a level that promotes vasoconstriction over vasodilation, thus altering cerebral blood flow increases in response to neuronal activity.
Collapse
Affiliation(s)
- Michaël Boily
- Department of Pharmacology and PhysiologyFaculty of MedicineUniversité de MontréalMontréalQuébecCanada
- Groupe de Recherche sur le Système Nerveux Central (GRSNC)Université de MontréalMontréalQuébecCanada
| | - Lin Li
- Department of Pharmacology and PhysiologyFaculty of MedicineUniversité de MontréalMontréalQuébecCanada
- Groupe de Recherche sur le Système Nerveux Central (GRSNC)Université de MontréalMontréalQuébecCanada
| | - Diane Vallerand
- Department of Pharmacology and PhysiologyFaculty of MedicineUniversité de MontréalMontréalQuébecCanada
- Groupe de Recherche sur le Système Nerveux Central (GRSNC)Université de MontréalMontréalQuébecCanada
- Centre interdisciplinaire de recherche sur le cerveau et l’apprentissage (CIRCA)Université de MontréalMontréalQuébecCanada
| | - Hélène Girouard
- Department of Pharmacology and PhysiologyFaculty of MedicineUniversité de MontréalMontréalQuébecCanada
- Groupe de Recherche sur le Système Nerveux Central (GRSNC)Université de MontréalMontréalQuébecCanada
- Centre interdisciplinaire de recherche sur le cerveau et l’apprentissage (CIRCA)Université de MontréalMontréalQuébecCanada
- Centre de Recherche de l’Institut de Gériatrie de MontréalMontréalQuébecCanada
| |
Collapse
|
18
|
Huber G, Ogrodnik M, Wenzel J, Stölting I, Huber L, Will O, Peschke E, Matschl U, Hövener JB, Schwaninger M, Jurk D, Raasch W. Telmisartan prevents high-fat diet-induced neurovascular impairments and reduces anxiety-like behavior. J Cereb Blood Flow Metab 2021; 41:2356-2369. [PMID: 33730932 PMCID: PMC8393307 DOI: 10.1177/0271678x211003497] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Angiotensin II receptor blockers (telmisartan) prevent rodents from diet-induced obesity and improve their metabolic status. Hyperglycemia and obesity are associated with reduced cerebral blood flow and neurovascular uncoupling which may lead to behavioral deficits. We wanted to know whether a treatment with telmisartan prevents these changes in obesity.We put young mice on high-fat diet and simultaneously treated them with telmisartan. At the end of treatment, we performed laser speckle imaging and magnetic resonance imaging to assess the effect on neurovascular coupling and cerebral blood flow. Different behavioral tests were used to investigate cognitive function.Mice developed diet-induced obesity and after 16, not 8 weeks of high-fat diet, however, the response to whisker pad stimulation was about 30% lower in obese compared to lean mice. Simultaneous telmisartan treatment increased the response again by 10% compared to obese mice. Moreover, telmisartan treatment normalized high-fat diet-induced reduction of cerebral blood flow and prevented a diet-induced anxiety-like behavior. In addition to that, telmisartan affects cellular senescence and string vessel formation in obesity.We conclude, that telmisartan protects against neurovascular unit impairments in a diet-induced obesity setting and may play a role in preventing obesity related cognitive deficits in Alzheimer's disease.
Collapse
Affiliation(s)
- Gianna Huber
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany.,CBBM (Centre for Brain, Behavior and Metabolism), University of Lübeck, Lübeck, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany
| | - Mikolaj Ogrodnik
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester MN, USA.,Ludwig Boltzmann Research Group Senescence and Healing of Wounds at LBI Trauma, Vienna, Austria
| | - Jan Wenzel
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany.,CBBM (Centre for Brain, Behavior and Metabolism), University of Lübeck, Lübeck, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany
| | - Ines Stölting
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany.,CBBM (Centre for Brain, Behavior and Metabolism), University of Lübeck, Lübeck, Germany
| | - Lukas Huber
- Section Biomedical Imaging, MOIN CC, Department of Radiology and Neuroradiology, UKSH, Kiel University, Kiel, Germany
| | - Olga Will
- Section Biomedical Imaging, MOIN CC, Department of Radiology and Neuroradiology, UKSH, Kiel University, Kiel, Germany
| | - Eva Peschke
- Section Biomedical Imaging, MOIN CC, Department of Radiology and Neuroradiology, UKSH, Kiel University, Kiel, Germany
| | - Urte Matschl
- Department Virus Immunology, Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Jan-Bernd Hövener
- Section Biomedical Imaging, MOIN CC, Department of Radiology and Neuroradiology, UKSH, Kiel University, Kiel, Germany
| | - Markus Schwaninger
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany.,CBBM (Centre for Brain, Behavior and Metabolism), University of Lübeck, Lübeck, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany
| | - Diana Jurk
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester MN, USA
| | - Walter Raasch
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany.,CBBM (Centre for Brain, Behavior and Metabolism), University of Lübeck, Lübeck, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Lübeck, Germany
| |
Collapse
|
19
|
A.S. Murad H, S. Al-Kayy S, A. Alkaabi T. Are the Newest Angiotensin Receptor Blockers Preferred than the Old Members in COVID-19 Patients with Cardiovascular Comorbidity? INT J PHARMACOL 2021. [DOI: 10.3923/ijp.2021.435.441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
20
|
Cuddy LK, Prokopenko D, Cunningham EP, Brimberry R, Song P, Kirchner R, Chapman BA, Hofmann O, Hide W, Procissi D, Hanania T, Leiser SC, Tanzi RE, Vassar R. Aβ-accelerated neurodegeneration caused by Alzheimer's-associated ACE variant R1279Q is rescued by angiotensin system inhibition in mice. Sci Transl Med 2021; 12:12/563/eaaz2541. [PMID: 32998969 DOI: 10.1126/scitranslmed.aaz2541] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 05/06/2020] [Indexed: 12/18/2022]
Abstract
Recent genome-wide association studies identified the angiotensin-converting enzyme gene (ACE) as an Alzheimer's disease (AD) risk locus. However, the pathogenic mechanism by which ACE causes AD is unknown. Using whole-genome sequencing, we identified rare ACE coding variants in AD families and investigated one, ACE1 R1279Q, in knockin (KI) mice. Similar to AD, ACE1 was increased in neurons, but not microglia or astrocytes, of KI brains, which became elevated further with age. Angiotensin II (angII) and angII receptor AT1R signaling were also increased in KI brains. Autosomal dominant neurodegeneration and neuroinflammation occurred with aging in KI hippocampus, which were absent in the cortex and cerebellum. Female KI mice exhibited greater hippocampal electroencephalograph disruption and memory impairment compared to males. ACE variant effects were more pronounced in female KI mice, suggesting a mechanism for higher AD risk in women. Hippocampal neurodegeneration was completely rescued by treatment with brain-penetrant drugs that inhibit ACE1 and AT1R. Although ACE variant-induced neurodegeneration did not depend on β-amyloid (Aβ) pathology, amyloidosis in 5XFAD mice crossed to KI mice accelerated neurodegeneration and neuroinflammation, whereas Aβ deposition was unchanged. KI mice had normal blood pressure and cerebrovascular functions. Our findings strongly suggest that increased ACE1/angII signaling causes aging-dependent, Aβ-accelerated selective hippocampal neuron vulnerability and female susceptibility, hallmarks of AD that have hitherto been enigmatic. We conclude that repurposed brain-penetrant ACE inhibitors and AT1R blockers may protect against AD.
Collapse
Affiliation(s)
- Leah K Cuddy
- Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Dmitry Prokopenko
- Genetics and Aging Unit and McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA
| | - Eric P Cunningham
- Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Ross Brimberry
- Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Peter Song
- Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Rory Kirchner
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Brad A Chapman
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Oliver Hofmann
- Department of Clinical Pathology, University of Melbourne, Victoria 3000, Melbourne, Australia
| | - Winston Hide
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Daniele Procissi
- Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | | | | | - Rudolph E Tanzi
- Genetics and Aging Unit and McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital, Boston, MA 02129, USA.
| | - Robert Vassar
- Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA. .,Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
21
|
Revi N, Rengan AK. Impact of dietary polyphenols on neuroinflammation-associated disorders. Neurol Sci 2021; 42:3101-3119. [PMID: 33988799 DOI: 10.1007/s10072-021-05303-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 05/05/2021] [Indexed: 12/21/2022]
Abstract
Neurodegenerative disorders like Alzheimer's, Parkinson's, and associated dementia typically originate with altered protein folding and aggregation of their β structures in the neurons. This self-aggregation leads to glial activation in the brain, causing neuroinflammation and leads to neuronal death. According to statistics provided by WHO, there are around 50 million people with dementia worldwide and every year, 10 million more cases are projected to increase. Also, around 5-8 percentage of people who are aged above 60 globally has dementia or associated disorders. Over 82 million in 2030 and 152 in 2050 are expected to have dementia. Most of these patients fall into low-middle-income countries which makes it even more essential to find an affordable and effective treatment method. Polyphenols of different origin are studied for their potential role as anti-neuro-inflammatory molecules. This review would summarize recent advances in three widely researched dietary polyphenols projected as potential therapeutic agents for disorders like Alzheimer's, Parkinson's, etc. They are Resveratrol, Catechins, and Tannins. The review would discuss the recent advances and challenges in using these polyphenols using specific examples as potential therapeutic agents against neuroinflammation associated disorders. An abstract of neuroinflammation-associated events and the effects by selected polyphenols.
Collapse
Affiliation(s)
- Neeraja Revi
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, India
| | - Aravind Kumar Rengan
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, India.
| |
Collapse
|
22
|
Saavedra JM. Angiotensin Receptor Blockers Are Not Just for Hypertension Anymore. Physiology (Bethesda) 2021; 36:160-173. [PMID: 33904788 DOI: 10.1152/physiol.00036.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Beyond blood pressure control, angiotensin receptor blockers reduce common injury mechanisms, decreasing excessive inflammation and protecting endothelial and mitochondrial function, insulin sensitivity, the coagulation cascade, immune responses, cerebrovascular flow, and cognition, properties useful to treat inflammatory, age-related, neurodegenerative, and metabolic disorders of many organs including brain and lung.
Collapse
Affiliation(s)
- Juan M Saavedra
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, District of Columbia
| |
Collapse
|
23
|
Ribeiro VT, Cordeiro TME, Filha RDS, Perez LG, Caramelli P, Teixeira AL, de Souza LC, Simões E Silva AC. Circulating Angiotensin-(1-7) Is Reduced in Alzheimer's Disease Patients and Correlates With White Matter Abnormalities: Results From a Pilot Study. Front Neurosci 2021; 15:636754. [PMID: 33897352 PMCID: PMC8063113 DOI: 10.3389/fnins.2021.636754] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 03/12/2021] [Indexed: 11/29/2022] Open
Abstract
Introduction Alzheimer’s disease (AD) is the leading cause of dementia worldwide. Despite the extensive research, its pathophysiology remains largely unelucidated. Currently, more attention is being given to the disease’s vascular and inflammatory aspects. In this context, the renin-angiotensin system (RAS) emerges as a credible player in AD pathogenesis. The RAS has multiple physiological functions, conducted by its two opposing axes: the classical, led by Angiotensin II (Ang II), and the alternative, driven by Angiotensin-(1–7) [Ang-(1–7)]. These peptides were shown to interact with AD pathology in animal studies, but evidence from humans is scarce. Only 20 studies dosed RAS molecules in AD patients’ bloodstream, none of which assessed both axes simultaneously. Therefore, we conducted a cross-sectional, case-control exploratory study to compare plasma levels of Ang II and Ang-(1–7) in AD patients vs. age-matched controls. Within each group, we searched for correlations between RAS biomarkers and measures from magnetic resonance imaging (MRI). Methods We evaluated patients with AD (n = 14) and aged-matched controls (n = 14). Plasma Ang II and Ang-(1–7) were dosed using ELISA. Brain MRI was performed in a 3 Tesla scan, and a three-dimensional T1-weighted volumetric sequence was obtained. Images were then processed by FreeSurfer to calculate: (1) white matter hypointensities (WMH) volume; (2) volumes of hippocampus, medial temporal cortex, and precuneus. Statistical analyses used non-parametrical tests (Mann-Whitney and Spearman). Results Ang-(1–7) levels in plasma were significantly lower in the AD patients than in controls [median (25th–75th percentiles)]: AD [101.5 (62.43–126.4)] vs. controls [209.3 (72–419.1)], p = 0.014. There was no significant difference in circulating Ang II. In the AD patients, but not in controls, there was a positive and significant correlation between Ang-(1–7) values and WMH volumes (Spearman’s rho = 0.56, p = 0.038). Ang-(1–7) did not correlate with cortical volumes in AD or in controls. Ang II did not correlate with any MRI variable in none of the groups. Conclusion If confirmed, our results strengthen the hypothesis that RAS alternative axis is downregulated in AD, and points to a possible interaction between Ang-(1–7) and cerebrovascular lesions in AD.
Collapse
Affiliation(s)
- Victor Teatini Ribeiro
- Laboratório Interdisciplinar de Investigação Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Thiago Macedo E Cordeiro
- Laboratório Interdisciplinar de Investigação Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Roberta da Silva Filha
- Laboratório Interdisciplinar de Investigação Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Lucas Giandoni Perez
- Laboratório Interdisciplinar de Investigação Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Paulo Caramelli
- Departamento de Clínica Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Antônio Lúcio Teixeira
- Neuropsychiatry Program and Immuno-Psychiatry Lab, Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Leonardo Cruz de Souza
- Laboratório Interdisciplinar de Investigação Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil.,Departamento de Clínica Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Ana Cristina Simões E Silva
- Laboratório Interdisciplinar de Investigação Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| |
Collapse
|
24
|
Attilio PJ, Snapper DM, Rusnak M, Isaac A, Soltis AR, Wilkerson MD, Dalgard CL, Symes AJ. Transcriptomic Analysis of Mouse Brain After Traumatic Brain Injury Reveals That the Angiotensin Receptor Blocker Candesartan Acts Through Novel Pathways. Front Neurosci 2021; 15:636259. [PMID: 33828448 PMCID: PMC8019829 DOI: 10.3389/fnins.2021.636259] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 02/19/2021] [Indexed: 12/30/2022] Open
Abstract
Traumatic brain injury (TBI) results in complex pathological reactions, where the initial lesion is followed by secondary inflammation and edema. Our laboratory and others have reported that angiotensin receptor blockers (ARBs) have efficacy in improving recovery from traumatic brain injury in mice. Treatment of mice with a subhypotensive dose of the ARB candesartan results in improved functional recovery, and reduced pathology (lesion volume, inflammation and gliosis). In order to gain a better understanding of the molecular mechanisms through which candesartan improves recovery after controlled cortical impact injury (CCI), we performed transcriptomic profiling on brain regions after injury and drug treatment. We examined RNA expression in the ipsilateral hippocampus, thalamus and hypothalamus at 3 or 29 days post injury (dpi) treated with either candesartan (0.1 mg/kg) or vehicle. RNA was isolated and analyzed by bulk mRNA-seq. Gene expression in injured and/or candesartan treated brain region was compared to that in sham vehicle treated mice in the same brain region to identify genes that were differentially expressed (DEGs) between groups. The most DEGs were expressed in the hippocampus at 3 dpi, and the number of DEGs reduced with distance and time from the lesion. Among pathways that were differentially expressed at 3 dpi after CCI, candesartan treatment altered genes involved in angiogenesis, interferon signaling, extracellular matrix regulation including integrins and chromosome maintenance and DNA replication. At 29 dpi, candesartan treatment reduced the expression of genes involved in the inflammatory response. Some changes in gene expression were confirmed in a separate cohort of animals by qPCR. Fewer DEGs were found in the thalamus, and only one in the hypothalamus at 3 dpi. Additionally, in the hippocampi of sham injured mice, 3 days of candesartan treatment led to the differential expression of 384 genes showing that candesartan in the absence of injury had a powerful impact on gene expression specifically in the hippocampus. Our results suggest that candesartan has broad actions in the brain after injury and affects different processes at acute and chronic times after injury. These data should assist in elucidating the beneficial effect of candesartan on recovery from TBI.
Collapse
Affiliation(s)
- Peter J. Attilio
- Graduate Program in Neuroscience, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Dustin M. Snapper
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Milan Rusnak
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Akira Isaac
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Anthony R. Soltis
- The American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Matthew D. Wilkerson
- The American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Clifton L. Dalgard
- The American Genome Center, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Aviva J. Symes
- Graduate Program in Neuroscience, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
25
|
Scheinman SB, Zaldua S, Dada A, Krochmaliuk K, Dye K, Marottoli FM, Thatcher GRJ, Tai LM. Systemic Candesartan Treatment Modulates Behavior, Synaptic Protein Levels, and Neuroinflammation in Female Mice That Express Human APOE4. Front Neurosci 2021; 15:628403. [PMID: 33642985 PMCID: PMC7902885 DOI: 10.3389/fnins.2021.628403] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 01/20/2021] [Indexed: 11/26/2022] Open
Abstract
Evidence suggests that angiotensin receptor blockers (ARBs) could be beneficial for Alzheimer’s disease (AD) patients independent of any effects on hypertension. However, studies in rodent models directly testing the activity of ARB treatment on behavior and AD-relevent pathology including neuroinflammation, Aβ levels, and cerebrovascular function, have produced mixed results. APOE4 is a major genetic risk factor for AD and has been linked to many of the same functions as those purported to be modulated by ARB treatment. Therefore, evaluating the effects of ARB treatment on behavior and AD-relevant pathology in mice that express human APOE4 could provide important information on whether to further develop ARBs for AD therapy. In this study, we treated female and male mice that express the human APOE4 gene in the absence (E4FAD−) or presence (E4FAD+) of high Aβ levels with the ARB prodrug candesartan cilexetil for a duration of 4 months. Compared to vehicle, candesartan treatment resulted in greater memory-relevant behavior and higher hippocampal presynaptic protein levels in female, but not male, E4FAD− and E4FAD+ mice. The beneficial effects of candesartan in female E4FAD− and E4FAD+ mice occurred in tandem with lower GFAP and Iba1 levels in the hippocampus, whereas there were no effects on markers of cerebrovascular function and Aβ levels. Collectively, these data imply that the effects of ARBs on AD-relevant pathology may be modulated in part by the interaction between APOE genotype and biological sex. Thus, the further development of ARBs could provide therapeutic options for targeting neuroinflammation in female APOE4 carriers.
Collapse
Affiliation(s)
- Sarah B Scheinman
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Steve Zaldua
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Adedoyin Dada
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Kateryna Krochmaliuk
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Katherine Dye
- UICentre, University of Illinois at Chicago, Chicago, IL, United States
| | - Felecia M Marottoli
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Gregory R J Thatcher
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, United States
| | - Leon M Tai
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
26
|
Ribeiro VT, de Souza LC, Simões E Silva AC. Renin-Angiotensin System and Alzheimer's Disease Pathophysiology: From the Potential Interactions to Therapeutic Perspectives. Protein Pept Lett 2020; 27:484-511. [PMID: 31886744 DOI: 10.2174/0929866527666191230103739] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 08/27/2019] [Accepted: 11/16/2019] [Indexed: 12/21/2022]
Abstract
New roles of the Renin-Angiotensin System (RAS), apart from fluid homeostasis and Blood Pressure (BP) regulation, are being progressively unveiled, since the discoveries of RAS alternative axes and local RAS in different tissues, including the brain. Brain RAS is reported to interact with pathophysiological mechanisms of many neurological and psychiatric diseases, including Alzheimer's Disease (AD). Even though AD is the most common cause of dementia worldwide, its pathophysiology is far from elucidated. Currently, no treatment can halt the disease course. Successive failures of amyloid-targeting drugs have challenged the amyloid hypothesis and increased the interest in the inflammatory and vascular aspects of AD. RAS compounds, both centrally and peripherally, potentially interact with neuroinflammation and cerebrovascular regulation. This narrative review discusses the AD pathophysiology and its possible interaction with RAS, looking forward to potential therapeutic approaches. RAS molecules affect BP, cerebral blood flow, neuroinflammation, and oxidative stress. Angiotensin (Ang) II, via angiotensin type 1 receptors may promote brain tissue damage, while Ang-(1-7) seems to elicit neuroprotection. Several studies dosed RAS molecules in AD patients' biological material, with heterogeneous results. The link between AD and clinical conditions related to classical RAS axis overactivation (hypertension, heart failure, and chronic kidney disease) supports the hypothesized role of this system in AD. Additionally, RAStargeting drugs as Angiotensin Converting Enzyme inhibitors (ACEis) and Angiotensin Receptor Blockers (ARBs) seem to exert beneficial effects on AD. Results of randomized controlled trials testing ACEi or ARBs in AD are awaited to elucidate whether AD-RAS interaction has implications on AD therapeutics.
Collapse
Affiliation(s)
- Victor Teatini Ribeiro
- Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Leonardo Cruz de Souza
- Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil.,Department of Internal Medicine, Service of Neurology, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Ana Cristina Simões E Silva
- Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| |
Collapse
|
27
|
Haghighi MM, Kakhki EG, Sato C, Ghani M, Rogaeva E. The Intersection between COVID-19, the Gene Family of ACE2 and Alzheimer's Disease. Neurosci Insights 2020; 15:2633105520975743. [PMID: 33283188 PMCID: PMC7686598 DOI: 10.1177/2633105520975743] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 11/03/2020] [Indexed: 01/08/2023] Open
Abstract
We reviewed factors that might influence COVID-19 outcomes (eg, neurological symptoms), including the link to Alzheimer's disease. Since the virus triggers COVID-19 infection through binding to ACE2, we focused on the ACE2 gene family, including ACE. Both ACE2 and ACE are involved in the renin-angiotensin system (RAS). In general, ACE causes inflammation and vasoconstriction, while ACE2 leads to anti-inflammation activity and vasodilation. The disturbed balance between these counter-regulatory pathways could influence susceptibility to COVID-19. Notably, dysregulation of the RAS-equilibrium contributes to Alzheimer's disease. Differences in the incidence and symptoms of COVID-19 in diverse populations could be attributed to variability in the human genome. For example, ACE and ACE2 variations could modify the outcome of COVID-19 in different populations. It would be important to conduct genome-wide studies to detect variants influencing COVID-19 presentation, with a special focus on variants affecting immune-related pathways and expression of RAS-related genes.
Collapse
Affiliation(s)
- Mahdi Montazer Haghighi
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - Erfan Ghani Kakhki
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada.,DisorDATA Analytics, Ottawa, ON, Canada
| | - Christine Sato
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | | | - Ekaterina Rogaeva
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada.,Division of Neurology, Department of Medicine, University of Toronto, Toronto, Canada
| |
Collapse
|
28
|
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
|
29
|
Kim JY, Barua S, Jeong YJ, Lee JE. Adiponectin: The Potential Regulator and Therapeutic Target of Obesity and Alzheimer's Disease. Int J Mol Sci 2020; 21:ijms21176419. [PMID: 32899357 PMCID: PMC7504582 DOI: 10.3390/ijms21176419] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 08/28/2020] [Accepted: 08/28/2020] [Indexed: 02/08/2023] Open
Abstract
Animal and human mechanistic studies have consistently shown an association between obesity and Alzheimer’s disease (AD). AD, a degenerative brain disease, is the most common cause of dementia and is characterized by the presence of extracellular amyloid beta (Aβ) plaques and intracellular neurofibrillary tangles disposition. Some studies have recently demonstrated that Aβ and tau cannot fully explain the pathophysiological development of AD and that metabolic disease factors, such as insulin, adiponectin, and antioxidants, are important for the sporadic onset of nongenetic AD. Obesity prevention and treatment can be an efficacious and safe approach to AD prevention. Adiponectin is a benign adipokine that sensitizes the insulin receptor signaling pathway and suppresses inflammation. It has been shown to be inversely correlated with adipose tissue dysfunction and may enhance the risk of AD because a range of neuroprotection adiponectin mechanisms is related to AD pathology alleviation. In this study, we summarize the recent progress that addresses the beneficial effects and potential mechanisms of adiponectin in AD. Furthermore, we review recent studies on the diverse medications of adiponectin that could possibly be related to AD treatment, with a focus on their association with adiponectin. A better understanding of the neuroprotection roles of adiponectin will help clarify the precise underlying mechanism of AD development and progression.
Collapse
Affiliation(s)
- Jong Youl Kim
- Department of Anatomy, Yonsei University College of Medicine, Seoul 120-752, Korea; (J.Y.K.); (S.B.); (Y.J.J.)
| | - Sumit Barua
- Department of Anatomy, Yonsei University College of Medicine, Seoul 120-752, Korea; (J.Y.K.); (S.B.); (Y.J.J.)
| | - Ye Jun Jeong
- Department of Anatomy, Yonsei University College of Medicine, Seoul 120-752, Korea; (J.Y.K.); (S.B.); (Y.J.J.)
| | - Jong Eun Lee
- Department of Anatomy, Yonsei University College of Medicine, Seoul 120-752, Korea; (J.Y.K.); (S.B.); (Y.J.J.)
- BK21 Plus Project for Medical Sciences, and Brain Research Institute, Yonsei University College of Medicine, Seoul 120-752, Korea
- Correspondence: ; Tel.: +82-2-2228-1646 (ext. 1659); Fax: +82-2-365-0700
| |
Collapse
|
30
|
Ahmed HA, Ishrat T. The Brain AT2R-a Potential Target for Therapy in Alzheimer's Disease and Vascular Cognitive Impairment: a Comprehensive Review of Clinical and Experimental Therapeutics. Mol Neurobiol 2020; 57:3458-3484. [PMID: 32533467 PMCID: PMC8109287 DOI: 10.1007/s12035-020-01964-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 05/28/2020] [Indexed: 10/24/2022]
Abstract
Dementia is a potentially avertable tragedy, currently considered among the top 10 greatest global health challenges of the twenty-first century. Dementia not only robs individuals of their dignity and independence, it also has a ripple effect that starts with the inflicted individual's family and projects to the society as a whole. The constantly growing number of cases, along with the lack of effective treatments and socioeconomic impact, poses a serious threat to the sustainability of our health care system. Hence, there is a worldwide effort to identify new targets for the treatment of Alzheimer's disease (AD), the leading cause of dementia. Due to its multifactorial etiology and the recent clinical failure of several novel amyloid-β (Aβ) targeting therapies, a comprehensive "multitarget" approach may be most appropriate for managing this condition. Interestingly, renin angiotensin system (RAS) modulators were shown to positively impact all the factors involved in the pathophysiology of dementia including vascular dysfunction, Aβ accumulation, and associated cholinergic deficiency, in addition to tau hyperphosphorylation and insulin derangements. Furthermore, for many of these drugs, the preclinical evidence is also supported by epidemiological data and/or preliminary clinical trials. The purpose of this review is to provide a comprehensive update on the major causes of dementia including the risk factors, current diagnostic criteria, pathophysiology, and contemporary treatment strategies. Moreover, we highlight the angiotensin II receptor type 2 (AT2R) as an effective drug target and present ample evidence supporting its potential role and clinical applications in cognitive impairment to encourage further investigation in the clinical setting.
Collapse
Affiliation(s)
- Heba A Ahmed
- Department of Anatomy and Neurobiology, College of Medicine, University of Tennessee Health Science Center, 855 Monroe Avenue, Wittenborg Bldg, Room-231, Memphis, TN, 38163, USA
| | - Tauheed Ishrat
- Department of Anatomy and Neurobiology, College of Medicine, University of Tennessee Health Science Center, 855 Monroe Avenue, Wittenborg Bldg, Room-231, Memphis, TN, 38163, USA.
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA.
- Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
31
|
Vaiserman A, Koliada A, Lushchak O. Neuroinflammation in pathogenesis of Alzheimer's disease: Phytochemicals as potential therapeutics. Mech Ageing Dev 2020; 189:111259. [PMID: 32450086 DOI: 10.1016/j.mad.2020.111259] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 05/02/2020] [Accepted: 05/02/2020] [Indexed: 02/07/2023]
Abstract
Accumulation of neurotoxic forms of amyloid-β proteins in senile plaques and hyperphosphorylated tau proteins in neurofibrillary tangles is a well-known pathophysiological hallmark of Alzheimer's disease (AD). However, clinical trials with drugs targeting amyloid-β and tau have failed to demonstrate efficacy in treating AD. All currently FDA-approved anti-AD drugs have symptomatic effects only and are not able to cure this disease. This makes necessary to search for alternative therapeutic targets. Accumulating evidence suggests that systemic inflammation and related vascular dysfunction play important etiological roles in AD and precede its clinical manifestation. Therefore, novel therapeutic modalities targeted at these pathophysiological components of AD are intensively developed now. Phytochemicals such as resveratrol, curcumin, quercetin, genistein and catechins are promising anti-AD therapeutics due to their ability to affect major pathogenetic mechanisms of AD, including oxidative stress, neuroinflammation and mitochondrial dysfunction. The implementation of innovative approaches for phytochemical delivery, including the nanotechnology-based ones which enable to significantly enhance their oral bioavailability, would likely provide an opportunity to address many challenges of conventional anti-AD therapies. In this review, roles of inflammation and vascular dysregulation in AD are described and phytobioactive compound-based treatment strategies for AD are discussed.
Collapse
Affiliation(s)
- Alexander Vaiserman
- Laboratory of Epigenetics, D.F. Chebotarev Institute of Gerontology, NAMS, 67 Vyshgorodska str., Kyiv, 04114, Ukraine.
| | - Alexander Koliada
- Laboratory of Epigenetics, D.F. Chebotarev Institute of Gerontology, NAMS, 67 Vyshgorodska str., Kyiv, 04114, Ukraine
| | - Oleh Lushchak
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenka str., Ivano-Frankivsk, 76018, Ukraine
| |
Collapse
|
32
|
Association between Use of Angiotensin-converting Enzyme Inhibitors or Angiotensin Receptor Blockers and Postoperative Delirium. Anesthesiology 2020; 133:119-132. [DOI: 10.1097/aln.0000000000003329] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Background
Angiotensin-converting enzyme inhibitors and angiotensin receptor blockers improve cognitive function. The authors therefore tested the primary hypothesis that preoperative use of angiotensin inhibitors is associated with less delirium in critical care patients. Post hoc, the association between postoperative use of angiotensin system inhibitors and delirium was assessed.
Methods
The authors conducted a single-site cohort study of adults admitted to Cleveland Clinic critical care units after noncardiac procedures between 2013 and 2018 who had at least one Confusion Assessment Method delirium assessment. Patients with preexisting dementia, Alzheimer’s disease or other cognitive decline, and patients who had neurosurgical procedures were excluded. For the primary analysis, the confounder-adjusted association between preoperative angiotensin inhibitor use and the incidence of postoperative delirium was assessed. Post hoc, the confounder-adjusted association between postoperative angiotensin system inhibitor use and the incidence of delirium was assessed.
Results
The incidence of delirium was 39% (551 of 1,396) among patients who were treated preoperatively with angiotensin system inhibitors and 39% (1,344 of 3,468) in patients who were not. The adjusted odds ratio of experiencing delirium during critical care was 0.98 (95% CI, 0.86 to 1.10; P = 0.700) for preoperative use of angiotensin system inhibitors versus control. Delirium was observed in 23% (100 of 440) of patients who used angiotensin system inhibitors postoperatively before intensive care discharge, and in 41% (1,795 of 4,424) of patients who did not (unadjusted P < 0.001). The confounder-adjusted odds ratio for experiencing delirium in patients who used angiotensin system inhibitors postoperatively was 0.55 (95% CI, 0.43 to 0.72; P < 0.001).
Conclusions
Preoperative use of angiotensin system inhibitors is not associated with reduced postoperative delirium. In contrast, treatment during intensive care was associated with lower odds of delirium. Randomized trials of postoperative angiotensin-converting enzymes inhibitors and angiotensin receptor blockers seem justified.
Editor’s Perspective
What We Already Know about This Topic
What This Article Tells Us That Is New
Collapse
|
33
|
Costa R, Tamascia ML, Sanches A, Moreira RP, Cunha TS, Nogueira MD, Casarini DE, Marcondes FK. Tactile stimulation of adult rats modulates hormonal responses, depression-like behaviors, and memory impairment induced by chronic mild stress: Role of angiotensin II. Behav Brain Res 2020; 379:112250. [DOI: 10.1016/j.bbr.2019.112250] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 09/17/2019] [Accepted: 09/17/2019] [Indexed: 12/19/2022]
|
34
|
Role of brain renin angiotensin system in neurodegeneration: An update. Saudi J Biol Sci 2020; 27:905-912. [PMID: 32127770 PMCID: PMC7042626 DOI: 10.1016/j.sjbs.2020.01.026] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 01/20/2020] [Accepted: 01/22/2020] [Indexed: 01/12/2023] Open
Abstract
Renin angiotensin system (RAS) is an endocrine system widely known for its physiological roles in electrolyte homeostasis, body fluid volume regulation and cardiovascular control in peripheral circulation. However, brain RAS is an independent form of RAS expressed locally in the brain, which is known to be involved in brain functions and disorders. There is strong evidence for a major involvement of excessive brain angiotensin converting enzyme (ACE)/Angiotensin II (Ang II)/Angiotensin type-1 receptor (AT-1R) axis in increased activation of oxidative stress, apoptosis and neuroinflammation causing neurodegeneration in several brain disorders. Numerous studies have demonstrated strong neuroprotective effects by blocking AT1R in these brain disorders. Additionally, the angiotensin converting enzyme 2 (ACE2)/Angiotensin (1–7)/Mas receptor (MASR), is another axis of brain RAS which counteracts the damaging effects of ACE/Ang II/AT1R axis on neurons in the brain. Thus, angiotensin II receptor blockers (ARBs) and activation of ACE2/Angiotensin (1–7)/MASR axis may serve as an exciting and novel method for neuroprotection in several neurodegenerative diseases. Here in this review article, we discuss the expression of RAS in the brain and highlight how altered RAS level may cause neurodegeneration. Understanding the pathophysiology of RAS and their links to neurodegeneration has enormous potential to identify potentially effective pharmacological tools to treat neurodegenerative diseases in the brain.
Collapse
|
35
|
Wang ZF, Li J, Ma C, Huang C, Li ZQ. Telmisartan ameliorates Aβ oligomer-induced inflammation via PPARγ/PTEN pathway in BV2 microglial cells. Biochem Pharmacol 2020; 171:113674. [DOI: 10.1016/j.bcp.2019.113674] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 10/16/2019] [Indexed: 12/18/2022]
|
36
|
Candesartan Neuroprotection in Rat Primary Neurons Negatively Correlates with Aging and Senescence: a Transcriptomic Analysis. Mol Neurobiol 2019; 57:1656-1673. [PMID: 31811565 DOI: 10.1007/s12035-019-01800-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 09/22/2019] [Indexed: 12/11/2022]
Abstract
Preclinical experiments and clinical trials demonstrated that angiotensin II AT1 receptor overactivity associates with aging and cellular senescence and that AT1 receptor blockers (ARBs) protect from age-related brain disorders. In a primary neuronal culture submitted to glutamate excitotoxicity, gene set enrichment analysis (GSEA) revealed expression of several hundred genes altered by glutamate and normalized by candesartan correlated with changes in expression in Alzheimer's patient's hippocampus. To further establish whether our data correlated with gene expression alterations associated with aging and senescence, we compared our global transcriptional data with additional published datasets, including alterations in gene expression in the neocortex and cerebellum of old mice, human frontal cortex after age of 40, gene alterations in the Werner syndrome, rodent caloric restriction, Ras and oncogene-induced senescence in fibroblasts, and to tissues besides the brain such as the muscle and kidney. The most significant and enriched pathways associated with aging and senescence were positively correlated with alterations in gene expression in glutamate-injured neurons and, conversely, negatively correlated when the injured neurons were treated with candesartan. Our results involve multiple genes and pathways, including CAV1, CCND1, CDKN1A, CHEK1, ICAM1, IL-1B, IL-6, MAPK14, PTGS2, SERPINE1, and TP53, encoding proteins associated with aging and senescence hallmarks, such as inflammation, oxidative stress, cell cycle and mitochondrial function alterations, insulin resistance, genomic instability including telomere shortening and DNA damage, and the senescent-associated secretory phenotype. Our results demonstrate that AT1 receptor blockade ameliorates central mechanisms of aging and senescence. Using ARBs for prevention and treatment of age-related disorders has important translational value.
Collapse
|
37
|
Mayburd AL, Koivogui M, Baranova A. Pharmacological signatures of the reduced incidence and the progression of cognitive decline in ageing populations suggest the protective role of beneficial polypharmacy. PLoS One 2019; 14:e0224315. [PMID: 31693707 PMCID: PMC6834256 DOI: 10.1371/journal.pone.0224315] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 10/10/2019] [Indexed: 12/13/2022] Open
Abstract
Preventive treatments for dementia are warranted. Here we show that utilization of certain combinations of prescription medications and supplements correlates with reduced rates of cognitive decline. More than 1,900 FDA-approved agents and supplements were collapsed into 53 mechanism-based groups and traced in electronic medical records (EMRs) for >50,000 patients. These mechanistic groups were aligned with the data presented in more than 300 clinical trials, then regression model was built to fit the signals from EMRs to clinical trial performance. While EMR signals of each single agents correlated with clinical performance relatively weakly, the signals produced by combinations of active compounds were highly correlated with the clinical trial performance (R = 0.93, p = 3.8 x10^-8). Higher ranking pharmacological modalities were traced in patient profiles as their combinations, producing protective complexity estimates reflecting degrees of exposure to beneficial polypharmacy. For each age strata, the higher was the protective complexity score, the lower was the prevalence of dementia, with maximized life-long effects for the highest regression score /diversity compositions. The connection was less strong in individuals already diagnosed with cognitive impairment. Confounder analysis confirmed an independent effect of protective complexity in multivariate context. A sub-cohort with lifelong odds of dementia decreased > 5-folds was identified; this sub-cohort should be studied in further details, including controlled clinical trials. In short, our study systematically explored combinatorial preventive treatment regimens for age-associated multi-morbidity, with an emphasis on neurodegeneration, and provided extensive evidence for their feasibility.
Collapse
Affiliation(s)
- Anatoly L. Mayburd
- Neurocombinatorix, Alexandria, Virginia, United States of America
- George Mason University, School of Systems Biology, Colgan Hall, MSN 3E1 George Mason University, Manassas, Virginia, United States of America
| | | | - Ancha Baranova
- George Mason University, School of Systems Biology, Colgan Hall, MSN 3E1 George Mason University, Manassas, Virginia, United States of America
- Research Centre for Medical Genetics, Moscow, Russia
| |
Collapse
|
38
|
Li T, Zhu J. Entanglement of CCR5 and Alzheimer's Disease. Front Aging Neurosci 2019; 11:209. [PMID: 31447666 PMCID: PMC6692443 DOI: 10.3389/fnagi.2019.00209] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 07/24/2019] [Indexed: 12/22/2022] Open
Abstract
Although the mechanisms of Alzheimer's disease are diverse and unclear, the past 20 years have witnessed the unprecedented development of the AD inflammation theory. As a key inflammatory receptor family, the C-C chemokine receptor family is a remarkable participant in the cause of Alzheimer's disease; of this family, CCR5 is the most widely studied. CCR5 is an essential entrance when HIV infects immune cells and is also involved in other inflammatory and immune activities. New evidence on the inevitably intertwined link between Alzheimer's disease and CCR5 indicates that CCR5 accelerates the development of Alzheimer's disease, and few studies disputed it. The role of CCR5 in Alzheimer's disease remains elusive. However, as the research progresses, this intricate relationship will gradually be uncovered.
Collapse
Affiliation(s)
- Tianwen Li
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology, Shanghai, China
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Jianhong Zhu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology, Shanghai, China
- Institutes of Brain Science, Shanghai, China
| |
Collapse
|
39
|
Ullah R, Khan M, Shah SA, Saeed K, Kim MO. Natural Antioxidant Anthocyanins-A Hidden Therapeutic Candidate in Metabolic Disorders with Major Focus in Neurodegeneration. Nutrients 2019; 11:E1195. [PMID: 31141884 PMCID: PMC6628002 DOI: 10.3390/nu11061195] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 05/23/2019] [Accepted: 05/24/2019] [Indexed: 12/15/2022] Open
Abstract
All over the world, metabolic syndrome constitutes severe health problems. Multiple factors have been reported in the pathogenesis of metabolic syndrome. Metabolic disorders result in reactive oxygen species (ROS) induced oxidative stress, playing a vital role in the development and pathogenesis of major health issues, including neurological disorders Alzheimer's disease (AD) Parkinson's disease (PD). Considerable increasing evidence indicates the substantial contribution of ROS-induced oxidative stress in neurodegenerative diseases. An imbalanced metabolism results in a defective antioxidant defense system, free radicals causing inflammation, cellular apoptosis, and tissue damage. Due to the annual increase in financial and social burdens, in addition to the adverse effects associated with available synthetic agents, treatment diversion from synthetic to natural approaches has occurred. Antioxidants are now being considered as convincing therapeutic agents against various neurodegenerative disorders. Therefore, medicinal herbs and fruits currently receive substantially more attention as commercial sources of antioxidants. In this review, we argue that ROS-targeted therapeutic interventions with naturally occurring antioxidant flavonoid, anthocyanin, and anthocyanin-loaded nanoparticles might be the ultimate treatment against devastating illnesses. Furthermore, we elucidate the hidden potential of the neuroprotective role of anthocyanins and anthocyanin-loaded nanoparticles in AD and PD neuropathies, which lack sufficient attention compared with other polyphenols, despite their strong antioxidant potential. Moreover, we address the need for future research studies of native anthocyanins and nano-based-anthocyanins, which will be helpful in developing anthocyanin treatments as therapeutic mitochondrial antioxidant drug-like regimens to delay or prevent the progression of neurodegenerative diseases, such as AD and PD.
Collapse
Affiliation(s)
- Rahat Ullah
- Division of Applied Life Science (BK 21), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea.
| | - Mehtab Khan
- Division of Applied Life Science (BK 21), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea.
| | - Shahid Ali Shah
- Division of Applied Life Science (BK 21), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea.
- Department of Chemistry, Sarhad University of Science & Information Technology (SUIT), Peshawar Khyber Pakhtunkhwa 25000, Pakistan.
| | - Kamran Saeed
- Division of Applied Life Science (BK 21), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea.
| | - Myeong Ok Kim
- Division of Applied Life Science (BK 21), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea.
| |
Collapse
|
40
|
Govindpani K, McNamara LG, Smith NR, Vinnakota C, Waldvogel HJ, Faull RL, Kwakowsky A. Vascular Dysfunction in Alzheimer's Disease: A Prelude to the Pathological Process or a Consequence of It? J Clin Med 2019; 8:E651. [PMID: 31083442 PMCID: PMC6571853 DOI: 10.3390/jcm8050651] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 04/29/2019] [Accepted: 05/06/2019] [Indexed: 12/19/2022] Open
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia. Despite decades of research following several theoretical and clinical lines, all existing treatments for the disorder are purely symptomatic. AD research has traditionally been focused on neuronal and glial dysfunction. Although there is a wealth of evidence pointing to a significant vascular component in the disease, this angle has been relatively poorly explored. In this review, we consider the various aspects of vascular dysfunction in AD, which has a significant impact on brain metabolism and homeostasis and the clearance of β-amyloid and other toxic metabolites. This may potentially precede the onset of the hallmark pathophysiological and cognitive symptoms of the disease. Pathological changes in vessel haemodynamics, angiogenesis, vascular cell function, vascular coverage, blood-brain barrier permeability and immune cell migration may be related to amyloid toxicity, oxidative stress and apolipoprotein E (APOE) genotype. These vascular deficits may in turn contribute to parenchymal amyloid deposition, neurotoxicity, glial activation and metabolic dysfunction in multiple cell types. A vicious feedback cycle ensues, with progressively worsening neuronal and vascular pathology through the course of the disease. Thus, a better appreciation for the importance of vascular dysfunction in AD may open new avenues for research and therapy.
Collapse
Affiliation(s)
- Karan Govindpani
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| | - Laura G McNamara
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| | - Nicholas R Smith
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| | - Chitra Vinnakota
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| | - Henry J Waldvogel
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| | - Richard Lm Faull
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| | - Andrea Kwakowsky
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
41
|
Xu X, Pan J, Li X, Cui Y, Mao Z, Wu B, Xu H, Zhou W, Liu Y. Inhibition of Methamphetamine Self-Administration and Reinstatement by Central Blockade of Angiotensin II Receptor in Rats. J Pharmacol Exp Ther 2019; 369:244-258. [PMID: 30867225 DOI: 10.1124/jpet.118.255729] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 03/04/2019] [Indexed: 02/06/2023] Open
Abstract
The molecular mechanism and treatment of methamphetamine (METH) use disorder remain unclear. The current study aimed to investigate the role of central angiotensin II receptor (ATR) in drug taking and seeking behavior associated with METH use disorder. The effect of an ATR type 1 (AT1R) antagonist, candesartan cilexetil, on the reinforcing and motivational effects of METH was first assessed using the animal model of METH self-administration (SA) and reinstatement. The levels of dopamine D2 receptor (D2R) and AT1R were subsequently examined. Furthermore, the present study determined the expression of microRNAs (miRNAs) by comparing METH SA, METH-yoked, and Saline-yoked groups. The target miRNAs were further overexpressed in the nucleus accumbens (NAc) via a lentivirus vector to investigate the effects of target miRNAs on METH SA maintained under a fixed ratio 1, progressive ratio, and cue/drug reinstatement of METH SA. The potential role of the AT1R-PLCβ-CREB signaling pathway was finally investigated. The results suggest that AT1R blockade effectively reduced METH SA and reinstatement, in conjunction with the counter-regulation of D2R and AT1R. A total of 17 miRNAs targeting Ang II in NAc were found to be associated with the voluntary intake of METH. Furthermore, overexpression of specific miR-219a-5p targeting AT1R-regulated METH SA and reinstatement. The AT1R-PLCβ-CREB signaling pathway was found to be associated with the effect of AT1R on the drug-taking and drug-seeking behavior involving METH use disorder.
Collapse
Affiliation(s)
- Xing Xu
- Ningbo University School of Medicine, Ningbo, Zhejiang, People's Republic of China (X.X., J.P., Z.M., B.W., W.Z., Y.L.); Ningbo Kangning Hospital, Ningbo, Zhejiang, People's Republic of China (X.L.); Ningbo Public Security Bureau Ningbo Anti-drug Office, Zhejiang, People's Republic of China (Y.C., H.X.); and Ningbo Addiction Research and Treatment Center, Zhejiang, People's Republic of China (W.Z.)
| | - Jian Pan
- Ningbo University School of Medicine, Ningbo, Zhejiang, People's Republic of China (X.X., J.P., Z.M., B.W., W.Z., Y.L.); Ningbo Kangning Hospital, Ningbo, Zhejiang, People's Republic of China (X.L.); Ningbo Public Security Bureau Ningbo Anti-drug Office, Zhejiang, People's Republic of China (Y.C., H.X.); and Ningbo Addiction Research and Treatment Center, Zhejiang, People's Republic of China (W.Z.)
| | - Xingxing Li
- Ningbo University School of Medicine, Ningbo, Zhejiang, People's Republic of China (X.X., J.P., Z.M., B.W., W.Z., Y.L.); Ningbo Kangning Hospital, Ningbo, Zhejiang, People's Republic of China (X.L.); Ningbo Public Security Bureau Ningbo Anti-drug Office, Zhejiang, People's Republic of China (Y.C., H.X.); and Ningbo Addiction Research and Treatment Center, Zhejiang, People's Republic of China (W.Z.)
| | - Yan Cui
- Ningbo University School of Medicine, Ningbo, Zhejiang, People's Republic of China (X.X., J.P., Z.M., B.W., W.Z., Y.L.); Ningbo Kangning Hospital, Ningbo, Zhejiang, People's Republic of China (X.L.); Ningbo Public Security Bureau Ningbo Anti-drug Office, Zhejiang, People's Republic of China (Y.C., H.X.); and Ningbo Addiction Research and Treatment Center, Zhejiang, People's Republic of China (W.Z.)
| | - Zijuan Mao
- Ningbo University School of Medicine, Ningbo, Zhejiang, People's Republic of China (X.X., J.P., Z.M., B.W., W.Z., Y.L.); Ningbo Kangning Hospital, Ningbo, Zhejiang, People's Republic of China (X.L.); Ningbo Public Security Bureau Ningbo Anti-drug Office, Zhejiang, People's Republic of China (Y.C., H.X.); and Ningbo Addiction Research and Treatment Center, Zhejiang, People's Republic of China (W.Z.)
| | - Boliang Wu
- Ningbo University School of Medicine, Ningbo, Zhejiang, People's Republic of China (X.X., J.P., Z.M., B.W., W.Z., Y.L.); Ningbo Kangning Hospital, Ningbo, Zhejiang, People's Republic of China (X.L.); Ningbo Public Security Bureau Ningbo Anti-drug Office, Zhejiang, People's Republic of China (Y.C., H.X.); and Ningbo Addiction Research and Treatment Center, Zhejiang, People's Republic of China (W.Z.)
| | - Huachong Xu
- Ningbo University School of Medicine, Ningbo, Zhejiang, People's Republic of China (X.X., J.P., Z.M., B.W., W.Z., Y.L.); Ningbo Kangning Hospital, Ningbo, Zhejiang, People's Republic of China (X.L.); Ningbo Public Security Bureau Ningbo Anti-drug Office, Zhejiang, People's Republic of China (Y.C., H.X.); and Ningbo Addiction Research and Treatment Center, Zhejiang, People's Republic of China (W.Z.)
| | - Wenhua Zhou
- Ningbo University School of Medicine, Ningbo, Zhejiang, People's Republic of China (X.X., J.P., Z.M., B.W., W.Z., Y.L.); Ningbo Kangning Hospital, Ningbo, Zhejiang, People's Republic of China (X.L.); Ningbo Public Security Bureau Ningbo Anti-drug Office, Zhejiang, People's Republic of China (Y.C., H.X.); and Ningbo Addiction Research and Treatment Center, Zhejiang, People's Republic of China (W.Z.)
| | - Yu Liu
- Ningbo University School of Medicine, Ningbo, Zhejiang, People's Republic of China (X.X., J.P., Z.M., B.W., W.Z., Y.L.); Ningbo Kangning Hospital, Ningbo, Zhejiang, People's Republic of China (X.L.); Ningbo Public Security Bureau Ningbo Anti-drug Office, Zhejiang, People's Republic of China (Y.C., H.X.); and Ningbo Addiction Research and Treatment Center, Zhejiang, People's Republic of China (W.Z.)
| |
Collapse
|
42
|
Pang C, Yang H, Hu B, Wang S, Chen M, Cohen DS, Chen HS, Jarrell JT, Carpenter KA, Rosin ER, Huang X. Identification and Analysis of Alzheimer's Candidate Genes by an Amplitude Deviation Algorithm. ACTA ACUST UNITED AC 2019; 9. [PMID: 31080696 PMCID: PMC6505709 DOI: 10.4172/2161-0460.1000460] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Background: Alzheimer’s disease (AD) is the most common form of senile dementia. However, its pathological mechanisms are not fully understood. In order to comprehend AD pathological mechanisms, researchers employed AD-related DNA microarray data and diverse computational algorithms. More efficient computational algorithms are needed to process DNA microarray data for identifying AD-related candidate genes. Methods: In this paper, we propose a specific algorithm that is based on the following observation: When an acrobat walks along a steel-wire, his/her body must have some swing; if the swing can be controlled, then the acrobat can maintain the body balance. Otherwise, the acrobat will fall. Based on this simple idea, we have designed a simple, yet practical, algorithm termed as the Amplitude Deviation Algorithm (ADA). Deviation, overall deviation, deviation amplitude, and 3δ are introduced to characterize ADA. Results: 52 candidate genes for AD have been identified via ADA. The implications for some of the AD candidate genes in AD pathogenesis have been discussed. Conclusions: Through the analysis of these AD candidate genes, we believe that AD pathogenesis may be related to the abnormality of signal transduction (AGTR1 and PTAFR), the decrease in protein transport capacity (COL5A2 (221729_at), COL5A2 (221730_at), COL4A1), the impairment of axon repair (CNR1), and the intracellular calcium dyshomeostasis (CACNB2, CACNA1E). However, their potential implication for AD pathology should be further validated by wet lab experiments as they were only identified by computation using ADA.
Collapse
Affiliation(s)
- Chaoyang Pang
- College of Computer Science, Sichuan Normal University, Chengdu, China
| | - Hualan Yang
- College of Mathematics and Software Science, Sichuan Normal University, Chengdu, China
| | - Benqiong Hu
- College of Management Science, Chengdu University of Technology, Chengdu, China
| | - Shipeng Wang
- College of Mathematics and Software Science, Sichuan Normal University, Chengdu, China
| | - Meixia Chen
- College of Mathematics and Software Science, Sichuan Normal University, Chengdu, China
| | - David S Cohen
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Hannah S Chen
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Juliet T Jarrell
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Kristy A Carpenter
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Eric R Rosin
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Xudong Huang
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| |
Collapse
|
43
|
Ibi D, Tsuchihashi A, Nomura T, Hiramatsu M. Involvement of GAT2/BGT-1 in the preventive effects of betaine on cognitive impairment and brain oxidative stress in amyloid β peptide-injected mice. Eur J Pharmacol 2019; 842:57-63. [DOI: 10.1016/j.ejphar.2018.10.037] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 10/13/2018] [Accepted: 10/25/2018] [Indexed: 12/17/2022]
|
44
|
Forrester SJ, Booz GW, Sigmund CD, Coffman TM, Kawai T, Rizzo V, Scalia R, Eguchi S. Angiotensin II Signal Transduction: An Update on Mechanisms of Physiology and Pathophysiology. Physiol Rev 2018; 98:1627-1738. [PMID: 29873596 DOI: 10.1152/physrev.00038.2017] [Citation(s) in RCA: 663] [Impact Index Per Article: 110.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The renin-angiotensin-aldosterone system plays crucial roles in cardiovascular physiology and pathophysiology. However, many of the signaling mechanisms have been unclear. The angiotensin II (ANG II) type 1 receptor (AT1R) is believed to mediate most functions of ANG II in the system. AT1R utilizes various signal transduction cascades causing hypertension, cardiovascular remodeling, and end organ damage. Moreover, functional cross-talk between AT1R signaling pathways and other signaling pathways have been recognized. Accumulating evidence reveals the complexity of ANG II signal transduction in pathophysiology of the vasculature, heart, kidney, and brain, as well as several pathophysiological features, including inflammation, metabolic dysfunction, and aging. In this review, we provide a comprehensive update of the ANG II receptor signaling events and their functional significances for potential translation into therapeutic strategies. AT1R remains central to the system in mediating physiological and pathophysiological functions of ANG II, and participation of specific signaling pathways becomes much clearer. There are still certain limitations and many controversies, and several noteworthy new concepts require further support. However, it is expected that rigorous translational research of the ANG II signaling pathways including those in large animals and humans will contribute to establishing effective new therapies against various diseases.
Collapse
Affiliation(s)
- Steven J Forrester
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - George W Booz
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Curt D Sigmund
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Thomas M Coffman
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Tatsuo Kawai
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Victor Rizzo
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Rosario Scalia
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Satoru Eguchi
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| |
Collapse
|
45
|
Diaz JR, Kim KJ, Brands MW, Filosa JA. Augmented astrocyte microdomain Ca 2+ dynamics and parenchymal arteriole tone in angiotensin II-infused hypertensive mice. Glia 2018; 67:551-565. [PMID: 30506941 DOI: 10.1002/glia.23564] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 10/22/2018] [Accepted: 10/22/2018] [Indexed: 11/09/2022]
Abstract
Hypertension is an important contributor to cognitive decline but the underlying mechanisms are unknown. Although much focus has been placed on the effect of hypertension on vascular function, less is understood of its effects on nonvascular cells. Because astrocytes and parenchymal arterioles (PA) form a functional unit (neurovascular unit), we tested the hypothesis that hypertension-induced changes in PA tone concomitantly increases astrocyte Ca2+ . We used cortical brain slices from 8-week-old mice to measure myogenic responses from pressurized and perfused PA. Chronic hypertension was induced in mice by 28-day angiotensin II (Ang II) infusion; PA resting tone and myogenic responses increased significantly. In addition, chronic hypertension significantly increased spontaneous Ca2+ events within astrocyte microdomains (MD). Similarly, a significant increase in astrocyte Ca2+ was observed during PA myogenic responses supporting enhanced vessel-to-astrocyte signaling. The transient potential receptor vanilloid 4 (TRPV4) channel, expressed in astrocyte processes in contact with blood vessels, namely endfeet, respond to hemodynamic stimuli such as increased pressure/flow. Supporting a role for TRPV4 channels in aberrant astrocyte Ca2+ dynamics in hypertension, cortical astrocytes from hypertensive mice showed augmented TRPV4 channel expression, currents and Ca2+ responses to the selective channel agonist GSK1016790A. In addition, pharmacological TRPV4 channel blockade or genetic deletion abrogated enhanced hypertension-induced increases in PA tone. Together, these data suggest chronic hypertension increases PA tone and Ca2+ events within astrocytes MD. We conclude that aberrant Ca2+ events in astrocyte constitute an early event toward the progression of cognitive decline.
Collapse
Affiliation(s)
| | - Ki Jung Kim
- Department of Physiology, Augusta University, Augusta, Georgia
| | | | | |
Collapse
|
46
|
Petek B, Villa-Lopez M, Loera-Valencia R, Gerenu G, Winblad B, Kramberger MG, Ismail MAM, Eriksdotter M, Garcia-Ptacek S. Connecting the brain cholesterol and renin-angiotensin systems: potential role of statins and RAS-modifying medications in dementia. J Intern Med 2018; 284:620-642. [PMID: 30264910 DOI: 10.1111/joim.12838] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Millions of people worldwide receive agents targeting the renin-angiotensin system (RAS) to treat hypertension or statins to lower cholesterol. The RAS and cholesterol metabolic pathways in the brain are autonomous from their systemic counterparts and are interrelated through the cholesterol metabolite 27-hydroxycholesterol (27-OHC). These systems contribute to memory and dementia pathogenesis through interference in the amyloid-beta cascade, vascular mechanisms, glucose metabolism, apoptosis, neuroinflammation and oxidative stress. Previous studies examining the relationship between these treatments and cognition and dementia risk have produced inconsistent results. Defining the blood-brain barrier penetration of these medications has been challenging, and the mechanisms of action on cognition are not clearly established. Potential biases are apparent in epidemiological and clinical studies, such as reverse epidemiology, indication bias, problems defining medication exposure, uncertain and changing doses, and inappropriate grouping of outcomes and medications. This review summarizes current knowledge of the brain cholesterol and RAS metabolism and the mechanisms by which these pathways affect neurodegeneration. The putative mechanisms of action of statins and medications inhibiting the RAS will be examined, together with prior clinical and animal studies on their effects on cognition. We review prior epidemiological studies, analysing their strengths and biases, and identify areas for future research. Understanding the pathophysiology of the brain cholesterol system and RAS and their links to neurodegeneration has enormous potential. In future, well-designed epidemiological studies could identify potential treatments for Alzheimer's disease (AD) amongst medications that are already in use for other indications.
Collapse
Affiliation(s)
- B Petek
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.,Department of Neurology, University Medical Centre, Ljubljana, Slovenia.,University of Ljubljana, Ljubljana, Slovenia
| | - M Villa-Lopez
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - R Loera-Valencia
- Center for Alzheimer Research, Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - G Gerenu
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.,Department of Neurosciences, Biodonostia Health Research Institute, San Sebastian, Spain.,Center for Networked Biomedical Research in Neurodegenerative Diseases, CIBERNED, Health Institute Carlos III, Ministry of Economy and Competitiveness, Madrid, Spain
| | - B Winblad
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.,Department of Geriatric Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - M G Kramberger
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.,Department of Neurology, University Medical Centre, Ljubljana, Slovenia.,University of Ljubljana, Ljubljana, Slovenia
| | - M-A-M Ismail
- Center for Alzheimer Research, Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.,Theme Neuro, Diseases of the Nervous System patient flow, Karolinska University Hospital, Huddinge, Sweden
| | - M Eriksdotter
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.,Department of Geriatric Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - S Garcia-Ptacek
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.,Department of Internal Medicine, Neurology Section, Södersjukhuset, Stockholm, Sweden
| |
Collapse
|
47
|
Angiotensin receptor (AT2R) agonist C21 prevents cognitive decline after permanent stroke in aged animals-A randomized double- blind pre-clinical study. Behav Brain Res 2018; 359:560-569. [PMID: 30296528 DOI: 10.1016/j.bbr.2018.10.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 09/10/2018] [Accepted: 10/04/2018] [Indexed: 12/20/2022]
Abstract
Post stroke cognitive impairment (PSCI) is an understudied, long-term complication of stroke, impacting nearly 30-40% of all stroke survivors. No cure is available once the cognitive deterioration manifests. To our knowledge, this is the first study to investigate the long-term effects of C21 treatment on the development of PSCI in aged animals. Treatments with C21 or vehicle were administered orally, 24 h post-stroke, and continued for 30 days. Outcome measures for sensorimotor and cognitive function were performed using a sequence of tests, all blindly conducted and assessed at baseline as well as at different time points post-stroke. Our findings demonstrate that the angiotensin receptor (AT2R) agonist C21 effectively prevents the development of PSCI in aged animals.
Collapse
|
48
|
Exploration of association of telmisartan with calf thymus DNA using a series of spectroscopic methodologies and theoretical calculation. J Mol Liq 2018. [DOI: 10.1016/j.molliq.2018.06.057] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
49
|
Ahmed HA, Ishrat T, Pillai B, Fouda AY, Sayed MA, Eldahshan W, Waller JL, Ergul A, Fagan SC. RAS modulation prevents progressive cognitive impairment after experimental stroke: a randomized, blinded preclinical trial. J Neuroinflammation 2018; 15:229. [PMID: 30103772 PMCID: PMC6090822 DOI: 10.1186/s12974-018-1262-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 07/29/2018] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND With the aging population, the prevalence and incidence of cerebrovascular disease will continue to rise, as well as the number of individuals with vascular cognitive impairment/dementia (VCID). No specific FDA-approved treatments for VCID exist. Although clinical evidence supports that angiotensin receptor blockers (ARBs) prevent cognitive decline in older adults, whether ARBs have a similar effect on VCID after stroke is unknown. Moreover, these agents reduce BP, which is undesirable in the acute stroke period, so we believe that giving C21 in this acute phase or delaying ARB administration would enable us to achieve the neurovascular benefits without the risk of unintended and potentially dangerous, acute BP lowering. METHODS The aim of our study was to determine the impact of candesartan (ARB) or compound-21 (an angiotensin type 2 receptor--AT2R--agonist) on long-term cognitive function post-stroke, in spontaneously hypertensive rats (SHRs). We hypothesized that AT2R stimulation, either directly with C21, or indirectly by blocking the angiotensin type 1 receptor (AT1R) with candesartan, initiated after stroke, would reduce cognitive impairment. Animals were subjected to a 60-min transient middle cerebral artery occlusion and randomly assigned to either saline/C21 monotherapy, for the full study duration (30 days), or given sequential therapy starting with saline/C21 (7 days) followed by candesartan for the remainder of the study (21 days). Outcome measures included sensorimotor/cognitive-function, amyloid-β determination, and histopathologic analyses. RESULTS Treatment with RAS modulators effectively preserved cognitive function, reduced cytotoxicity, and prevented chronic-reactive microgliosis in SHRs, post-stroke. These protective effects were apparent even when treatment was delayed up to 7 days post-stroke and were independent of blood pressure and β-amyloid accumulation. CONCLUSION Collectively, our findings demonstrate that RAS modulators effectively prevent cognitive impairment after stroke, even when treatment is delayed.
Collapse
Affiliation(s)
- Heba A. Ahmed
- Program in Clinical and Experimental Therapeutics, Charlie Norwood VA Medical Center and University of Georgia College of Pharmacy, HM Bldg., 1120 15th St, Augusta, GA 30912 USA
| | - Tauheed Ishrat
- Department of Anatomy and Neurobiology, College of Medicine, Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN USA
| | - Bindu Pillai
- Program in Clinical and Experimental Therapeutics, Charlie Norwood VA Medical Center and University of Georgia College of Pharmacy, HM Bldg., 1120 15th St, Augusta, GA 30912 USA
| | - Abdelrahman Y. Fouda
- Program in Clinical and Experimental Therapeutics, Charlie Norwood VA Medical Center and University of Georgia College of Pharmacy, HM Bldg., 1120 15th St, Augusta, GA 30912 USA
| | - Mohammed A. Sayed
- Program in Clinical and Experimental Therapeutics, Charlie Norwood VA Medical Center and University of Georgia College of Pharmacy, HM Bldg., 1120 15th St, Augusta, GA 30912 USA
| | - Wael Eldahshan
- Program in Clinical and Experimental Therapeutics, Charlie Norwood VA Medical Center and University of Georgia College of Pharmacy, HM Bldg., 1120 15th St, Augusta, GA 30912 USA
| | - Jennifer L. Waller
- Department of Biostatistics and Epidemiology, Augusta University, Augusta, GA USA
| | - Adviye Ergul
- Program in Clinical and Experimental Therapeutics, Charlie Norwood VA Medical Center and University of Georgia College of Pharmacy, HM Bldg., 1120 15th St, Augusta, GA 30912 USA
- Department of Physiology, Augusta University, Augusta, GA USA
| | - Susan C. Fagan
- Program in Clinical and Experimental Therapeutics, Charlie Norwood VA Medical Center and University of Georgia College of Pharmacy, HM Bldg., 1120 15th St, Augusta, GA 30912 USA
- Department of Neurology, Augusta University, Augusta, GA USA
| |
Collapse
|
50
|
Salmani H, Hosseini M, Beheshti F, Baghcheghi Y, Sadeghnia HR, Soukhtanloo M, Shafei MN, Khazaei M. Angiotensin receptor blocker, losartan ameliorates neuroinflammation and behavioral consequences of lipopolysaccharide injection. Life Sci 2018; 203:161-170. [DOI: 10.1016/j.lfs.2018.04.033] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 04/11/2018] [Accepted: 04/19/2018] [Indexed: 02/06/2023]
|