1
|
Yasmin S, Ashique S, Taj T, Garg A, Das J, Shorog E, Bhui U, Pal R, Selim S, Panigrahy UP, Begum N, Islam A, Ansari MY. The role of ACE inhibitors and ARBs in preserving cognitive function via hypertension Management: A critical Update. Brain Res 2024; 1850:149400. [PMID: 39681155 DOI: 10.1016/j.brainres.2024.149400] [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: 07/15/2024] [Revised: 11/27/2024] [Accepted: 12/10/2024] [Indexed: 12/18/2024]
Abstract
Hypertension poses a significant risk to cognition-related disorders like dementia. As the global population ages, age-related neurological illnesses such as Alzheimer's disease are becoming increasingly prevalent. The primary hypertension treatments, angiotensin receptor blockers, and angiotensin-converting enzyme inhibitors, exhibit neuroprotective properties. However, observational studies suggest that they may independently contribute to cognitive decline and dementia. Some of these medications have shown promise in reducing cognitive impairment and amyloid buildup in Alzheimer's models. While direct comparisons between the two drug classes are limited, angiotensin receptor blockers have been associated with less brain shrinkage, lower dementia incidence, and slower cognitive decline compared to angiotensin-converting enzyme inhibitors. Both types of medications can influence cognition by passing the blood-brain barrier, with angiotensin receptor blockers potentially offering superior neuroprotective effects due to their selective blockade of the angiotensin type 1 receptor.
Collapse
Affiliation(s)
- Sabina Yasmin
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - Sumel Ashique
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India.
| | - Tahreen Taj
- Department of Pharmacology, Yenepoya Pharmacy college and research centre, Yenepoya (Deemed to be) university, Mangalore 575018 , India
| | - Ashish Garg
- Guru Ramdas Khalsa Institute of Science and Technology, Pharmacy, Jabalpur, M.P, 483001, India
| | - Joy Das
- Department of Pharmacology, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab-144411, India
| | - Eman Shorog
- Clinical Pharmacy Department, College of Pharmacy, King Khalid University, Abha, 62529, Saudi Arabia
| | - Utpal Bhui
- Department of Pharmacology, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab-144411, India
| | - Radheshyam Pal
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India
| | - Samy Selim
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 72388, Saudi Arabia
| | - Uttam Prasad Panigrahy
- Department of Pharmaceutical Analysis, Faculty of Pharmaceutical Science, Assam down town University, SankarMadhab Path,Gandhi Nagar, Panikhaiti, Guwahati, Assam 781026, India
| | - Naseem Begum
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University Abha-62529, Saudi Arabia
| | - Anas Islam
- Faculty of Pharmacy, Integral University, Lucknow, 226026, Uttar Pradesh, India
| | - Mohammad Yousuf Ansari
- MM college of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana 133207, India.
| |
Collapse
|
2
|
Ali NH, Al‐Kuraishy HM, Al‐Gareeb AI, Alnaaim SA, Alexiou A, Papadakis M, Khalifa AA, Saad HM, Batiha GE. Neprilysin inhibitors and risk of Alzheimer's disease: A future perspective. J Cell Mol Med 2024; 28:e17993. [PMID: 37847125 PMCID: PMC10826440 DOI: 10.1111/jcmm.17993] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 09/15/2023] [Accepted: 10/05/2023] [Indexed: 10/18/2023] Open
Abstract
Alzheimer's disease (AD) is a heterogeneous neurodegenerative disease with multifaceted neuropathological disorders. AD is characterized by intracellular accumulation of phosphorylated tau proteins and extracellular deposition of amyloid beta (Aβ). Various protease enzymes, including neprilysin (NEP), are concerned with the degradation and clearance of Aβ. Indeed, a defective neuronal clearance pathway due to the dysfunction of degradation enzymes might be a possible mechanism for the accumulation of Aβ and subsequent progression of AD neuropathology. NEP is one of the most imperative metalloproteinase enzymes involved in the clearance of Aβ. This review aimed to highlight the possible role of NEP inhibitors in AD. The combination of sacubitril and valsartan which is called angiotensin receptor blocker and NEP inhibitor (ARNI) may produce beneficial and deleterious effects on AD neuropathology. NEP inhibitors might increase the risk of AD by the inhibition of Aβ clearance, and increase brain bradykinin (BK) and natriuretic peptides (NPs), which augment the pathogenesis of AD. These verdicts come from animal model studies, though they may not be applied to humans. However, clinical studies revealed promising safety findings regarding the use of ARNI. Moreover, NEP inhibition increases various neuroprotective peptides involved in inflammation, glucose homeostasis and nerve conduction. Also, NEP inhibitors may inhibit dipeptidyl peptidase 4 (DPP4) expression, ameliorating insulin and glucagon-like peptide 1 (GLP-1) levels. These findings proposed that NEP inhibitors may have a protective effect against AD development by increasing GLP-1, neuropeptide Y (NPY) and substance P, and deleterious effects by increasing brain BK. Preclinical and clinical studies are recommended in this regard.
Collapse
Affiliation(s)
- Naif H. Ali
- Department of Internal Medicine, Medical CollegeNajran UniversityNajranSaudi Arabia
| | - Hayder M. Al‐Kuraishy
- Department of Clinical Pharmacology and Medicine, College of MedicineMustansiriyah UniversityBaghdadIraq
| | - Ali I. Al‐Gareeb
- Department of Clinical Pharmacology and Medicine, College of MedicineMustansiriyah UniversityBaghdadIraq
| | - Saud A. Alnaaim
- Clinical Neurosciences Department, College of MedicineKing Faisal UniversityHofufSaudi Arabia
| | - Athanasios Alexiou
- Department of Science and EngineeringNovel Global Community Educational FoundationHebershamNew South WalesAustralia
- AFNP MedWienAustria
| | - Marios Papadakis
- Department of Surgery IIUniversity Hospital Witten‐Herdecke, University of Witten‐HerdeckeWuppertalGermany
| | - Asmaa A. Khalifa
- Department of Pharmacology and Therapeutics, Faculty of PharmacyPharos University in AlexandriaAlexandriaEgypt
| | - Hebatallah M. Saad
- Department of Pathology, Faculty of Veterinary MedicineMatrouh UniversityMatrouhEgypt
| | - Gaber El‐Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary MedicineDamanhour UniversityDamanhourAlBeheiraEgypt
| |
Collapse
|
3
|
Reveret L, Leclerc M, Emond V, Tremblay C, Loiselle A, Bourassa P, Bennett DA, Hébert SS, Calon F. Higher angiotensin-converting enzyme 2 (ACE2) levels in the brain of individuals with Alzheimer's disease. Acta Neuropathol Commun 2023; 11:159. [PMID: 37784209 PMCID: PMC10544218 DOI: 10.1186/s40478-023-01647-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 08/30/2023] [Indexed: 10/04/2023] Open
Abstract
Cognitive decline due to Alzheimer's disease (AD) is frequent in the geriatric population, which has been disproportionately affected by the COVID-19 pandemic. In this study, we investigated the levels of angiotensin-converting enzyme 2 (ACE2), a regulator of the renin-angiotensin system and the main entry receptor of SARS-CoV-2 in host cells, in postmortem parietal cortex samples from two independent AD cohorts, totalling 142 persons. Higher concentrations of ACE2 protein (p < 0.01) and mRNA (p < 0.01) were found in individuals with a neuropathological diagnosis of AD compared to age-matched healthy control subjects. Brain levels of soluble ACE2 were inversely associated with cognitive scores (p = 0.02) and markers of pericytes (PDGFRβ, p = 0.02 and ANPEP, p = 0.007), but positively correlated with concentrations of soluble amyloid-β peptides (Aβ) (p = 0.01) and insoluble phospho-tau (S396/404, p = 0.002). However, no significant differences in ACE2 were observed in the 3xTg-AD mouse model of tau and Aβ neuropathology. Results from immunofluorescence and Western blots showed that ACE2 protein is predominantly localized in microvessels in the mouse brain whereas it is more frequently found in neurons in the human brain. The present data suggest that higher levels of soluble ACE2 in the human brain may contribute to AD, but their role in CNS infection by SARS-CoV-2 remains unclear.
Collapse
Affiliation(s)
- Louise Reveret
- Faculty of Pharmacy, Laval University, Quebec, QC, Canada
- CHU de Quebec Research Center, 2705, Boulevard Laurier, Room T2-05, Québec, QC, G1V 4G2, Canada
| | - Manon Leclerc
- Faculty of Pharmacy, Laval University, Quebec, QC, Canada
- CHU de Quebec Research Center, 2705, Boulevard Laurier, Room T2-05, Québec, QC, G1V 4G2, Canada
| | - Vincent Emond
- CHU de Quebec Research Center, 2705, Boulevard Laurier, Room T2-05, Québec, QC, G1V 4G2, Canada
| | - Cyntia Tremblay
- CHU de Quebec Research Center, 2705, Boulevard Laurier, Room T2-05, Québec, QC, G1V 4G2, Canada
| | - Andréanne Loiselle
- CHU de Quebec Research Center, 2705, Boulevard Laurier, Room T2-05, Québec, QC, G1V 4G2, Canada
| | - Philippe Bourassa
- Faculty of Pharmacy, Laval University, Quebec, QC, Canada
- CHU de Quebec Research Center, 2705, Boulevard Laurier, Room T2-05, Québec, QC, G1V 4G2, Canada
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Sébastien S Hébert
- CHU de Quebec Research Center, 2705, Boulevard Laurier, Room T2-05, Québec, QC, G1V 4G2, Canada
- Faculty of Medicine, Laval University, Quebec, QC, Canada
| | - Frédéric Calon
- Faculty of Pharmacy, Laval University, Quebec, QC, Canada.
- CHU de Quebec Research Center, 2705, Boulevard Laurier, Room T2-05, Québec, QC, G1V 4G2, Canada.
| |
Collapse
|
4
|
Puranik N, Yadav D, Song M. Advancements in the Application of Nanomedicine in Alzheimer's Disease: A Therapeutic Perspective. Int J Mol Sci 2023; 24:14044. [PMID: 37762346 PMCID: PMC10530821 DOI: 10.3390/ijms241814044] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/11/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease that affects most people worldwide. AD is a complex central nervous system disorder. Several drugs have been designed to cure AD, but with low success rates. Because the blood-brain and blood-cerebrospinal fluid barriers are two barriers that protect the central nervous system, their presence has severely restricted the efficacy of many treatments that have been studied for AD diagnosis and/or therapy. The use of nanoparticles for the diagnosis and treatment of AD is the focus of an established and rapidly developing field of nanomedicine. Recent developments in nanomedicine have made it possible to effectively transport drugs to the brain. However, numerous obstacles remain to the successful use of nanomedicines in clinical settings for AD treatment. Furthermore, given the rapid advancement in nanomedicine therapeutics, better outcomes for patients with AD can be anticipated. This article provides an overview of recent developments in nanomedicine using different types of nanoparticles for the management and treatment of AD.
Collapse
Affiliation(s)
| | | | - Minseok Song
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea; (N.P.); (D.Y.)
| |
Collapse
|
5
|
Nassan M, Daghlas I, Piras IS, Rogalski E, Reus LM, Pijnenburg Y, Cuddy LK, Saxena R, Mesulam MM, Huentelman M. Evaluating the association between genetically proxied ACE inhibition and dementias. Alzheimers Dement 2023; 19:3894-3901. [PMID: 37023267 DOI: 10.1002/alz.13062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 03/03/2023] [Accepted: 03/08/2023] [Indexed: 04/08/2023]
Abstract
INTRODUCTION Angiotensin-converting enzyme (ACE) has been implicated in the metabolism of amyloid beta; however, the causal effect of ACE inhibition on risk of Alzheimer's disease (AD) dementia and other common dementias is largely unknown. METHODS We examined the causal association of genetically proxied ACE inhibition with four types of dementias using a two-sample Mendelian randomization (MR) approach. RESULTS Genetically proxied ACE inhibition was associated with increased risk of AD dementia (odds ratio per one standard deviation reduction in serum ACE [95% confidence interval]; 1.07 [1.04-1.10], P = 5 × 10-07 ) and frontotemporal dementia (1.16 [1.04-1.29], P = 0.01) but not with Lewy body dementia or vascular dementia (P > 0.05). These findings were independently replicated and remained consistent in sensitivity analyses. DISCUSSION This comprehensive MR study provided genetic evidence for an association between ACE inhibition and the risk for AD and frontotemporal dementias. These results should encourage further studies of the neurocognitive effects of ACE inhibition. HIGHLIGHTS This study evaluated genetically proxied angiotensin-converting enzyme (ACE) inhibition association with dementias. The results suggest an association between ACE inhibition and Alzheimer's disease. The results suggest an association between ACE inhibition and frontotemporal dementia. Those associations can be interpreted as potentially causal.
Collapse
Affiliation(s)
- Malik Nassan
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University, Chicago, Illinois, USA
| | - Iyas Daghlas
- Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Ignazio S Piras
- Neurogenomics Division, Translational Genomics Research Institute, Tgen, Phoenix, Arizona, USA
| | - Emily Rogalski
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University, Chicago, Illinois, USA
| | - Lianne M Reus
- Center for Neurobehavioral Genetics, University of California, Los Angeles, California, USA
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Yolande Pijnenburg
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Leah K Cuddy
- Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Richa Saxena
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - M-Marsel Mesulam
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University, Chicago, Illinois, USA
| | - Matt Huentelman
- Neurogenomics Division, Translational Genomics Research Institute, Tgen, Phoenix, Arizona, USA
| |
Collapse
|
6
|
Thong EHE, Quek EJW, Loo JH, Yun CY, Teo YN, Teo YH, Leow AST, Li TYW, Sharma VK, Tan BYQ, Yeo LLL, Chong YF, Chan MY, Sia CH. Acute Myocardial Infarction and Risk of Cognitive Impairment and Dementia: A Review. BIOLOGY 2023; 12:1154. [PMID: 37627038 PMCID: PMC10452707 DOI: 10.3390/biology12081154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 08/05/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023]
Abstract
Cognitive impairment (CI) shares common cardiovascular risk factors with acute myocardial infarction (AMI), and is increasingly prevalent in our ageing population. Whilst AMI is associated with increased rates of CI, CI remains underreported and infrequently identified in patients with AMI. In this review, we discuss the evidence surrounding AMI and its links to dementia and CI, including pathophysiology, risk factors, management and interventions. Vascular dysregulation plays a major role in CI, with atherosclerosis, platelet activation, microinfarcts and perivascular inflammation resulting in neurovascular unit dysfunction, disordered homeostasis and a dysfunctional neurohormonal response. This subsequently affects perfusion pressure, resulting in enlarged periventricular spaces and hippocampal sclerosis. The increased platelet activation seen in coronary artery disease (CAD) can also result in inflammation and amyloid-β protein deposition which is associated with Alzheimer's Dementia. Post-AMI, reduced blood pressure and reduced left ventricular ejection fraction can cause chronic cerebral hypoperfusion, cerebral infarction and failure of normal circulatory autoregulatory mechanisms. Patients who undergo coronary revascularization (percutaneous coronary intervention or bypass surgery) are at increased risk for post-procedure cognitive impairment, though whether this is related to the intervention itself or underlying cardiovascular risk factors is debated. Mortality rates are higher in dementia patients with AMI, and post-AMI CI is more prevalent in the elderly and in patients with post-AMI heart failure. Medical management (antiplatelet, statin, renin-angiotensin system inhibitors, cardiac rehabilitation) can reduce the risk of post-AMI CI; however, beta-blockers may be associated with functional decline in patients with existing CI. The early identification of those with dementia or CI who present with AMI is important, as subsequent tailoring of management strategies can potentially improve outcomes as well as guide prognosis.
Collapse
Affiliation(s)
- Elizabeth Hui En Thong
- Internal Medicine Residency, National University Health System, Singapore 119074, Singapore; (E.H.E.T.); (Y.H.T.); (A.S.T.L.)
| | - Ethan J. W. Quek
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; (E.J.W.Q.); (J.H.L.); (Y.N.T.); (V.K.S.); (B.Y.Q.T.); (L.L.L.Y.); (M.Y.C.)
| | - Jing Hong Loo
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; (E.J.W.Q.); (J.H.L.); (Y.N.T.); (V.K.S.); (B.Y.Q.T.); (L.L.L.Y.); (M.Y.C.)
| | - Choi-Ying Yun
- Department of Cardiology, National University Heart Centre Singapore, Singapore 119074, Singapore; (C.-Y.Y.); (T.Y.W.L.)
| | - Yao Neng Teo
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; (E.J.W.Q.); (J.H.L.); (Y.N.T.); (V.K.S.); (B.Y.Q.T.); (L.L.L.Y.); (M.Y.C.)
| | - Yao Hao Teo
- Internal Medicine Residency, National University Health System, Singapore 119074, Singapore; (E.H.E.T.); (Y.H.T.); (A.S.T.L.)
| | - Aloysius S. T. Leow
- Internal Medicine Residency, National University Health System, Singapore 119074, Singapore; (E.H.E.T.); (Y.H.T.); (A.S.T.L.)
| | - Tony Y. W. Li
- Department of Cardiology, National University Heart Centre Singapore, Singapore 119074, Singapore; (C.-Y.Y.); (T.Y.W.L.)
| | - Vijay K. Sharma
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; (E.J.W.Q.); (J.H.L.); (Y.N.T.); (V.K.S.); (B.Y.Q.T.); (L.L.L.Y.); (M.Y.C.)
- Division of Neurology, Department of Medicine, National University Hospital, Singapore 119074, Singapore;
| | - Benjamin Y. Q. Tan
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; (E.J.W.Q.); (J.H.L.); (Y.N.T.); (V.K.S.); (B.Y.Q.T.); (L.L.L.Y.); (M.Y.C.)
- Division of Neurology, Department of Medicine, National University Hospital, Singapore 119074, Singapore;
| | - Leonard L. L. Yeo
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; (E.J.W.Q.); (J.H.L.); (Y.N.T.); (V.K.S.); (B.Y.Q.T.); (L.L.L.Y.); (M.Y.C.)
- Division of Neurology, Department of Medicine, National University Hospital, Singapore 119074, Singapore;
| | - Yao Feng Chong
- Division of Neurology, Department of Medicine, National University Hospital, Singapore 119074, Singapore;
| | - Mark Y. Chan
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; (E.J.W.Q.); (J.H.L.); (Y.N.T.); (V.K.S.); (B.Y.Q.T.); (L.L.L.Y.); (M.Y.C.)
- Department of Cardiology, National University Heart Centre Singapore, Singapore 119074, Singapore; (C.-Y.Y.); (T.Y.W.L.)
| | - Ching-Hui Sia
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; (E.J.W.Q.); (J.H.L.); (Y.N.T.); (V.K.S.); (B.Y.Q.T.); (L.L.L.Y.); (M.Y.C.)
- Department of Cardiology, National University Heart Centre Singapore, Singapore 119074, Singapore; (C.-Y.Y.); (T.Y.W.L.)
| |
Collapse
|
7
|
Yang MH, Ho TC, Chang CC, Su YS, Yuan CH, Chuang KP, Tyan YC. Utilizing Proteomic Approaches to Uncover the Neuroprotective Effects of ACE Inhibitors: Implications for Alzheimer's Disease Treatment. Molecules 2023; 28:5938. [PMID: 37630190 PMCID: PMC10459293 DOI: 10.3390/molecules28165938] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/30/2023] [Accepted: 07/18/2023] [Indexed: 08/27/2023] Open
Abstract
Two types of angiotensin-converting enzyme (ACE) inhibitors, lisinopril and benazepril HCl, were tested in neuroblastoma cells and found to upregulate low-density lipoprotein-receptor-related protein 1B (LRP1B) and 14-3-3 protein zeta/delta. Additionally, benazepril HCl was found to increase the expression of calreticulin. The upregulation of these proteins by ACE inhibitors may contribute to the amelioration of cognitive deficits in Alzheimer's disease/dementia, as well as the clinically observed deceleration of functional decline in Alzheimer's patients. This discovery suggests that the supplementation of ACE inhibitors may promote neuronal cell survival independently of their antihypertensive effect. Overall, these findings indicate that ACE inhibitors may be a promising avenue for developing effective treatments for Alzheimer's disease.
Collapse
Affiliation(s)
- Ming-Hui Yang
- Division of General & Digestive Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Tzu-Chuan Ho
- Department of Medical Imaging and Radiological Sciences, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chin-Chuan Chang
- Department of Nuclear Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- School of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yuh-Shan Su
- Department of Medical Imaging and Radiological Sciences, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Cheng-Hui Yuan
- Mass Spectrometry Laboratory, Department of Chemistry, National University of Singapore, Singapore 119077, Singapore
| | - Kuo-Pin Chuang
- School of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- International Degree Program in Animal Vaccine Technology, International College, National Pingtung University of Science and Technology, Pingtung 912, Taiwan
- Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung 912, Taiwan
- Companion Animal Research Center, National Pingtung University of Science and Technology, Pingtung 912, Taiwan
| | - Yu-Chang Tyan
- Department of Medical Imaging and Radiological Sciences, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Nuclear Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung 912, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Center for Tropical Medicine and Infectious Disease Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
8
|
Waigi EW, Webb RC, Moss MA, Uline MJ, McCarthy CG, Wenceslau CF. Soluble and insoluble protein aggregates, endoplasmic reticulum stress, and vascular dysfunction in Alzheimer's disease and cardiovascular diseases. GeroScience 2023; 45:1411-1438. [PMID: 36823398 PMCID: PMC10400528 DOI: 10.1007/s11357-023-00748-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 01/28/2023] [Indexed: 02/25/2023] Open
Abstract
Dementia refers to a particular group of symptoms characterized by difficulties with memory, language, problem-solving, and other thinking skills that affect a person's ability to perform everyday activities. Alzheimer's disease (AD) is the most common form of dementia, affecting about 6.2 million Americans aged 65 years and older. Likewise, cardiovascular diseases (CVDs) are a major cause of disability and premature death, impacting 126.9 million adults in the USA, a number that increases with age. Consequently, CVDs and cardiovascular risk factors are associated with an increased risk of AD and cognitive impairment. They share important age-related cardiometabolic and lifestyle risk factors, that make them among the leading causes of death. Additionally, there are several premises and hypotheses about the mechanisms underlying the association between AD and CVD. Although AD and CVD may be considered deleterious to health, the study of their combination constitutes a clinical challenge, and investigations to understand the mechanistic pathways for the cause-effect and/or shared pathology between these two disease constellations remains an active area of research. AD pathology is propagated by the amyloid β (Aβ) peptides. These peptides give rise to small, toxic, and soluble Aβ oligomers (SPOs) that are nonfibrillar, and it is their levels that show a robust correlation with the extent of cognitive impairment. This review will elucidate the interplay between the effects of accumulating SPOs in AD and CVDs, the resulting ER stress response, and their role in vascular dysfunction. We will also address the potential underlying mechanisms, including the possibility that SPOs are among the causes of vascular injury in CVD associated with cognitive decline. By revealing common mechanistic underpinnings of AD and CVD, we hope that novel experimental therapeutics can be designed to reduce the burden of these devastating diseases. Graphical abstract Alzheimer's disease (AD) pathology leads to the release of Aβ peptides, and their accumulation in the peripheral organs has varying effects on various components of the cardiovascular system including endoplasmic reticulum (ER) stress and vascular damage. Image created with BioRender.com.
Collapse
Affiliation(s)
- Emily W Waigi
- Cardiovascular Translational Research Cententer (CTRC), Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
| | - R Clinton Webb
- Cardiovascular Translational Research Cententer (CTRC), Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
- Biomedical Engineering Program, Univeristy of South Carolina, Columbia, SC, USA
| | - Melissa A Moss
- Biomedical Engineering Program, Univeristy of South Carolina, Columbia, SC, USA
- Department of Chemical Engineering, University of South Carolina, Columbia, SC, USA
| | - Mark J Uline
- Biomedical Engineering Program, Univeristy of South Carolina, Columbia, SC, USA
- Department of Chemical Engineering, University of South Carolina, Columbia, SC, USA
| | - Cameron G McCarthy
- Cardiovascular Translational Research Cententer (CTRC), Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
- Biomedical Engineering Program, Univeristy of South Carolina, Columbia, SC, USA
| | - Camilla Ferreira Wenceslau
- Cardiovascular Translational Research Cententer (CTRC), Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA.
- Biomedical Engineering Program, Univeristy of South Carolina, Columbia, SC, USA.
| |
Collapse
|
9
|
Kujovic M, Lipka T, Zalman M, Baumann L, Jänner M, Baumann B. Treatment of hypertension and obstructive sleep apnea counteracts cognitive decline in common neurocognitive disorders in diagnosis-related patterns. Sci Rep 2023; 13:7556. [PMID: 37160982 PMCID: PMC10169815 DOI: 10.1038/s41598-023-33701-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 04/18/2023] [Indexed: 05/11/2023] Open
Abstract
The aim of this study was to investigate the effect of arterial hypertension (AH) and of obstructive sleep apnea (OSA) on cognitive course in the neurocognitive disorder (NCD) cohort RIFADE which enrolled patients with NCD due to Alzheimer's disease (AD), vascular NCD (vNCD), and mixed NCD (AD + vNCD = mNCD). Multiple risk factors (RF), including AH and OSA, that contribute to the development of various kinds of dementia have been identified in previous studies. Studies that observed AH lacked investigation of long-term effects and did not isolate it from other RF. Studies involving OSA as a risk factor did not include participants with all stages of NCD. 126 subjects were screened for AH and OSA. Repeated cognitive measurements were performed with the DemTect as primary outcome and the clock drawing test as secondary outcome measure. 90 patients had AH (71.4%) and 40 patients had OSA (31.7%). RF-status had a significant effect on cognitive outcome in models with RF as single factors (AH p = 0.027, OSA p < 0.001), a 2-factor analysis with AH × OSA (AH as main factor p = 0.027) as well as a model including the 3 factors AH × OSA × diagnosis (p = 0.038). Similarly, a 3-factor model was significant for the clock-drawing test, whereas single factor-models remained insignificant. AH and OSA appear to be risk factors in common NCD and cognitive decline can be mitigated by treatment of these RF.
Collapse
Affiliation(s)
- Milenko Kujovic
- Department of Psychiatry and Psychotherapy, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany.
- Department of Neuropsychiatry, Centre for Neurology and Neuropsychiatry, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany.
| | - Tim Lipka
- Department of Psychiatry and Psychotherapy, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Mark Zalman
- Department of Psychiatry and Psychotherapy, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Leonie Baumann
- Department of Mental Health, University Hospital of Münster, Munster, Germany
| | - Michaela Jänner
- Department of Psychiatry and Psychotherapy, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Bruno Baumann
- Department of Mental Health, University Hospital of Münster, Munster, Germany
| |
Collapse
|
10
|
Arosio B, Calvani R, Ferri E, Coelho-Junior HJ, Carandina A, Campanelli F, Ghiglieri V, Marzetti E, Picca A. Sarcopenia and Cognitive Decline in Older Adults: Targeting the Muscle-Brain Axis. Nutrients 2023; 15:nu15081853. [PMID: 37111070 PMCID: PMC10142447 DOI: 10.3390/nu15081853] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 04/09/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Declines in physical performance and cognition are commonly observed in older adults. The geroscience paradigm posits that a set of processes and pathways shared among age-associated conditions may also serve as a molecular explanation for the complex pathophysiology of physical frailty, sarcopenia, and cognitive decline. Mitochondrial dysfunction, inflammation, metabolic alterations, declines in cellular stemness, and altered intracellular signaling have been observed in muscle aging. Neurological factors have also been included among the determinants of sarcopenia. Neuromuscular junctions (NMJs) are synapses bridging nervous and skeletal muscle systems with a relevant role in age-related musculoskeletal derangement. Patterns of circulating metabolic and neurotrophic factors have been associated with physical frailty and sarcopenia. These factors are mostly related to disarrangements in protein-to-energy conversion as well as reduced calorie and protein intake to sustain muscle mass. A link between sarcopenia and cognitive decline in older adults has also been described with a possible role for muscle-derived mediators (i.e., myokines) in mediating muscle-brain crosstalk. Herein, we discuss the main molecular mechanisms and factors involved in the muscle-brain axis and their possible implication in cognitive decline in older adults. An overview of current behavioral strategies that allegedly act on the muscle-brain axis is also provided.
Collapse
Affiliation(s)
- Beatrice Arosio
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| | - Riccardo Calvani
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, 00168 Rome, Italy
- Department of Geriatrics and Orthopedics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Evelyn Ferri
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Hélio José Coelho-Junior
- Department of Geriatrics and Orthopedics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Angelica Carandina
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| | - Federica Campanelli
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Veronica Ghiglieri
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, 00168 Rome, Italy
- San Raffaele University, 00168 Rome, Italy
| | - Emanuele Marzetti
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, 00168 Rome, Italy
- Department of Geriatrics and Orthopedics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Anna Picca
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, 00168 Rome, Italy
- Department of Medicine and Surgery, LUM University, 70100 Casamassima, Italy
| |
Collapse
|
11
|
Ma K, Zheng ZR, Meng Y. Pathogenesis of Chronic Kidney Disease Is Closely Bound up with Alzheimer's Disease, Especially via the Renin-Angiotensin System. J Clin Med 2023; 12:jcm12041459. [PMID: 36835994 PMCID: PMC9966558 DOI: 10.3390/jcm12041459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/01/2023] [Accepted: 02/07/2023] [Indexed: 02/15/2023] Open
Abstract
Chronic kidney disease (CKD) is a clinical syndrome secondary to the definitive change in function and structure of the kidney, which is characterized by its irreversibility and slow and progressive evolution. Alzheimer's disease (AD) is characterized by the extracellular accumulation of misfolded β-amyloid (Aβ) proteins into senile plaques and the formation of neurofibrillary tangles (NFTs) containing hyperphosphorylated tau. In the aging population, CKD and AD are growing problems. CKD patients are prone to cognitive decline and AD. However, the connection between CKD and AD is still unclear. In this review, we take the lead in showing that the development of the pathophysiology of CKD may also cause or exacerbate AD, especially the renin-angiotensin system (RAS). In vivo studies had already shown that the increased expression of angiotensin-converting enzyme (ACE) produces a positive effect in aggravating AD, but ACE inhibitors (ACEIs) have protective effects against AD. Among the possible association of risk factors in CKD and AD, we mainly discuss the RAS in the systemic circulation and the brain.
Collapse
Affiliation(s)
- Ke Ma
- The First Affiliated Hospital of Jinan University, Guangzhou 510000, China
| | - Zi-Run Zheng
- The First Affiliated Hospital of Jinan University, Guangzhou 510000, China
| | - Yu Meng
- The First Affiliated Hospital of Jinan University, Guangzhou 510000, China
- Central Laboratory, The Fifth Affiliated Hospital of Jinan University, Heyuan 517000, China
- Institute of Nephrology, Jinan University, Guangzhou 510000, China
- Correspondence:
| |
Collapse
|
12
|
Bueno V, Frasca D. Mini-review: Angiotensin- converting enzyme 1 (ACE1) and the impact for diseases such as Alzheimer's disease, sarcopenia, cancer, and COVID-19. FRONTIERS IN AGING 2023; 4:1117502. [PMID: 36756193 PMCID: PMC9899811 DOI: 10.3389/fragi.2023.1117502] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 01/11/2023] [Indexed: 01/24/2023]
Abstract
Ageing has been associated with comorbidities, systemic low-grade of inflammation, and immunosenescence. Hypertension is the most common morbidity and anti-hypertensives are used for more than 50%. Angiotensin-converting enzyme 1 inhibitors (ACEi) and angiotensin II receptor blockers (ARB) control blood pressure but also seem to play a role in comorbidities such as Alzheimer's disease, sarcopenia and cancer. The impact of anti-hypertensives in comorbidities is due to the expression of renin-angiotensin system (RAS) in several tissues and body fluids. Angiotensin-converting enzyme 1 (ACE1) has been linked to oxidative stress, metabolism, and inflammation. The levels and activity of ACE1 are under genetic control and polymorphisms have been correlated with susceptibility to Alzheimer's disease. In addition, some results found that ACEi and ARB users present delayed cognitive decline and reduced risk of dementia. Regarding to sarcopenia, RAS has been linked to the catabolic and anabolic pathways for muscle mass maintenance. In some studies, older adults using ACEi were highly benefited by exercise training. In cancer, RAS and its products have been shown to play a role since their inhibition in animal models modulates tumor microenvironment and improves the delivery of chemotherapy drugs. Clinically, the incidence of colorectal cancer is reduced in patients using ACEi and ARB. During the pandemic COVID-19 it was found that ACE2 receptor plays a role in the entry of SARS-CoV-2 into the host cell. ACE1 genotypes have been linked to an increased risk for COVID-19 and severe disease. In some studies COVID-19 patients taking ARB or ACEi presented better outcome.
Collapse
Affiliation(s)
- Valquiria Bueno
- Department of Microbiology Immunology and Parasitology, UNIFESP Federal University of São Paulo, São Paulo, Brazil
| | - Daniela Frasca
- Department of Immunology, University of Miami, Miami, FL, United States
| |
Collapse
|
13
|
Louise R, Manon L, Vincent E, Andréanne L, Philippe B, Cyntia T, Bennett DA, Sébastien H, Frédéric C. Higher Angiotensin I Converting Enzyme 2 (ACE2) levels in the brain of individuals with Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.17.524254. [PMID: 36711734 PMCID: PMC9882134 DOI: 10.1101/2023.01.17.524254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a major cause of death in the elderly. Cognitive decline due to Alzheimer's disease (AD) is frequent in the geriatric population disproportionately affected by the COVID-19 pandemic. Interestingly, central nervous system (CNS) manifestations have been reported in SARS-CoV-2-infected patients. In this study, we investigated the levels of Angiotensin I Converting Enzyme 2 (ACE2), the main entry receptor of SARS-COV-2 in cells, in postmortem parietal cortex samples from two independent AD cohorts, totalling 142 persons. Higher concentrations of ACE2 protein and mRNA were found in individuals with a neuropathological diagnosis of AD compared to age-matched healthy control subjects. Brain levels of soluble ACE2 were inversely associated with cognitive scores (p = 0.02), markers of pericytes (PDGFRβ, p=0.02 and ANPEP, p = 0.007) and caveolin1 (p = 0.03), but positively correlated with soluble amyloid-β peptides (Aβ) concentrations (p = 0.01) and insoluble phospho- tau (S396/404, p = 0.002). No significant differences in ACE2 were observed in the 3xTgAD mouse model of tau and Aβ neuropathology. Results from immunofluorescence and Western blots showed that ACE2 protein is mainly localized in neurons in the human brain but predominantly in microvessels in the mouse brain. The present data show that an AD diagnosis is associated with higher levels of soluble ACE2 in the human brain, which might contribute to a higher risk of CNS SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Reveret Louise
- Faculty of pharmacy, Laval University, Quebec, QC, Canada
- CHU de Quebec Research Center, Quebec, QC, Canada
| | - Leclerc Manon
- Faculty of pharmacy, Laval University, Quebec, QC, Canada
- CHU de Quebec Research Center, Quebec, QC, Canada
| | | | | | - Bourassa Philippe
- Faculty of pharmacy, Laval University, Quebec, QC, Canada
- CHU de Quebec Research Center, Quebec, QC, Canada
| | | | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Hébert Sébastien
- CHU de Quebec Research Center, Quebec, QC, Canada
- Faculty of medicine, Laval University, Quebec, QC, Canada
| | - Calon Frédéric
- Faculty of pharmacy, Laval University, Quebec, QC, Canada
- CHU de Quebec Research Center, Quebec, QC, Canada
| |
Collapse
|
14
|
Ghalayini J, Boulianne GL. Deciphering mechanisms of action of ACE inhibitors in neurodegeneration using Drosophila models of Alzheimer's disease. Front Neurosci 2023; 17:1166973. [PMID: 37113150 PMCID: PMC10126366 DOI: 10.3389/fnins.2023.1166973] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 03/17/2023] [Indexed: 04/29/2023] Open
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disorder for which there is no cure. Recently, several studies have reported a significant reduction in the incidence and progression of dementia among some patients receiving antihypertensive medications such as angiotensin-converting enzyme inhibitors (ACE-Is) and angiotensin receptor blockers (ARBs). Why these drugs are beneficial in some AD patients and not others is unclear although it has been shown to be independent of their role in regulating blood pressure. Given the enormous and immediate potential of ACE-Is and ARBs for AD therapeutics it is imperative that we understand how they function. Recently, studies have shown that ACE-Is and ARBs, which target the renin angiotensin system in mammals, are also effective in suppressing neuronal cell death and memory defects in Drosophila models of AD despite the fact that this pathway is not conserved in flies. This suggests that the beneficial effects of these drugs may be mediated by distinct and as yet, identified mechanisms. Here, we discuss how the short lifespan and ease of genetic manipulations available in Drosophila provide us with a unique and unparalleled opportunity to rapidly identify the targets of ACE-Is and ARBs and evaluate their therapeutic effectiveness in robust models of AD.
Collapse
Affiliation(s)
- Judy Ghalayini
- Program in Developmental and Stem Cell Biology, Peter Gilgin Center for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Gabrielle L. Boulianne
- Program in Developmental and Stem Cell Biology, Peter Gilgin Center for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- *Correspondence: Gabrielle L. Boulianne,
| |
Collapse
|
15
|
Nagy A, Májer R, Csikai E, Dobos A, Süvegh G, Csiba L. The Correlation between Two Angiotensin-Converting Enzyme Inhibitor's Concentrations and Cognition. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:14375. [PMID: 36361252 PMCID: PMC9654214 DOI: 10.3390/ijerph192114375] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/23/2022] [Accepted: 10/30/2022] [Indexed: 06/16/2023]
Abstract
Both lisinopril and enalapril are angiotensin-converting enzyme (ACE) drugs and widely used in the treatment of hypertension. Enalapril does not cross the blood-brain barrier, but lisinopril is centrally active. Our goal was to find out if there was a link between the actual concentration of ACE inhibitors and cognition and if there was a detectable difference between the two types of ACE inhibitors. Asymptomatic, non-treated patients were diagnosed by screening and the hypertension was confirmed by ambulatory blood pressure monitoring (ABPM). A battery of cognitive tests was used to assess the impact of randomly assigning participants to receive either lisinopril or enalapril. All neurocognitive functions were measured, especially the most affected by conditions of compromised perfusion pressures, such as hypertension, which are attention and executive functions. The lisinopril concentration showed a significant inverse correlation with mosaic test (coeff. = -0.5779) and seemed to have a significant negative effect on perceptual motor skills (coeff. = -0.5779), complex attention (coeff. = -0.5104) and learning (coeff. = -0.5202). Compared with enalapril, lisinopril is less successful in improving the components of cognitive functions.
Collapse
Affiliation(s)
- Attila Nagy
- Department of Health Informatics, Faculty of Health Sciences, University of Debrecen, 4032 Debrecen, Hungary
| | - Réka Májer
- Department of Neurology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Enikő Csikai
- Doctoral School of Health Sciences, Institute of Behavioral Science, Faculty of General Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Adrienn Dobos
- Department of Forensic Toxicology, Hungarian Institute for Forensic Sciences, 1903 Budapest, Hungary
| | - Gábor Süvegh
- Department of Forensic Toxicology, Hungarian Institute for Forensic Sciences, 1903 Budapest, Hungary
| | - László Csiba
- Department of Neurology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
16
|
Cognitive dysfunction in SLE: An understudied clinical manifestation. J Autoimmun 2022; 132:102911. [PMID: 36127204 DOI: 10.1016/j.jaut.2022.102911] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 08/31/2022] [Indexed: 11/22/2022]
Abstract
Neuropsychiatric lupus (NPSLE) is a debilitating manifestation of SLE which occurs in a majority of SLE patients and has a variety of clinical manifestations. In the central nervous system, NPSLE may result from ischemia or penetration of inflammatory mediators and neurotoxic antibodies through the blood brain barrier (BBB). Here we focus on cognitive dysfunction (CD) as an NPSLE manifestation; it is common, underdiagnosed, and without specific therapy. For a very long time, clinicians ignored cognitive dysfunction and researchers who might be interested in the question struggled to find an approach to understanding mechanisms for this manifestation. Recent years, however, propelled by a more patient-centric approach to disease, have seen remarkable progress in our understanding of CD pathogenesis. This has been enabled through the use of novel imaging modalities and numerous mouse models. Overall, these studies point to a pivotal role of an impaired BBB and microglial activation in leading to neuronal injury. These insights suggest potential therapeutic modalities and make possible clinical trials for cognitive impairment.
Collapse
|
17
|
Correa BHM, Becari L, Peliky Fontes MA, Simões-e-Silva AC, Kangussu LM. Involvement of the Renin-Angiotensin System in Stress: State of the Art and Research Perspectives. Curr Neuropharmacol 2022; 20:1212-1228. [PMID: 34554902 PMCID: PMC9886820 DOI: 10.2174/1570159x19666210719142300] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 06/19/2021] [Accepted: 07/09/2021] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Along with other canonical systems, the renin-angiotensin system (RAS) has shown important roles in stress. This system is a complex regulatory proteolytic cascade composed of various enzymes, peptides, and receptors. Besides the classical (ACE/Ang II/AT1 receptor) and the counter-regulatory (ACE2/Ang-(1-7)/Mas receptor) RAS axes, evidence indicates that nonclassical components, including Ang III, Ang IV, AT2 and AT4, can also be involved in stress. OBJECTIVE AND METHODS This comprehensive review summarizes the current knowledge on the participation of RAS components in different adverse environmental stimuli stressors, including air jet stress, cage switch stress, restraint stress, chronic unpredictable stress, neonatal isolation stress, and post-traumatic stress disorder. RESULTS AND CONCLUSION In general, activation of the classical RAS axis potentiates stress-related cardiovascular, endocrine, and behavioral responses, while the stimulation of the counter-regulatory axis attenuates these effects. Pharmacological modulation in both axes is optimistic, offering promising perspectives for stress-related disorders treatment. In this regard, angiotensin-converting enzyme inhibitors and angiotensin receptor blockers are potential candidates already available since they block the classical axis, activate the counter-regulatory axis, and are safe and efficient drugs.
Collapse
Affiliation(s)
- Bernardo H. M. Correa
- Department of Morphology, Biological Sciences Institute, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil;
| | - Luca Becari
- Department of Morphology, Biological Sciences Institute, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil;
| | - Marco Antônio Peliky Fontes
- Department of Physiology & Biophysics - Biological Sciences Institute, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil;
| | - Ana Cristina Simões-e-Silva
- Department of Pediatrics, Faculty of Medicine, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Lucas M. Kangussu
- Department of Morphology, Biological Sciences Institute, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil; ,Address correspondence to this author at the Department of Morphology, Biological Sciences Institute – Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil; Tel: (+55-31) 3409-2772; E-mail:
| |
Collapse
|
18
|
Gouveia F, Camins A, Ettcheto M, Bicker J, Falcão A, Cruz MT, Fortuna A. Targeting brain Renin-Angiotensin System for the prevention and treatment of Alzheimer's disease: Past, present and future. Ageing Res Rev 2022; 77:101612. [PMID: 35346852 DOI: 10.1016/j.arr.2022.101612] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 02/09/2022] [Accepted: 03/16/2022] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is a well-known neurodegenerative disease characterized by the presence of two main hallmarks - Tau hyperphosphorylation and Aβ deposits. Notwithstanding, in the last few years the scientific evidence about the drivers of AD have been changing and nowadays age-related vascular alterations and several cardiovascular risk factors have been shown to trigger the development of AD. In this context, drugs targeting the Renin Angiotensin System (RAS), commonly used for the treatment of hypertension, are evidencing a high potential to delay AD development due to their action on brain RAS. Indeed, the ACE 1/Ang II/AT1R axis is believed to be upregulated in AD and to be responsible for deleterious effects such as increased oxidative stress, neuroinflammation, blood-brain barrier (BBB) hyperpermeability, astrocytes dysfunction and a decrease in cerebral blood flow. In contrast, the alternative axis - ACE 1/Ang II/AT2R; ACE 2/Ang (1-7)/MasR; Ang IV/ AT4R(IRAP) - seems to counterbalance the deleterious effects of the principal axis and to exert beneficial effects on memory and cognition. Accordingly, retrospective studies demonstrate a reduced risk of developing AD among people taking RAS medication as well as several in vitro and in vivo pre-clinical studies as it is herein critically reviewed. In this review, we first revise, at a glance, the pathophysiology of AD focused on its classic hallmarks. Secondly, an overview about the impact of the RAS on the pathophysiology of AD is also provided, focused on their four essential axes ACE 1/Ang II/AT2R; ACE 2/Ang (1-7)/MasR; Ang IV/ AT4R(IRAP) and ACE 1/Ang II/AT1R. Finally, the therapeutic potential of available drugs targeting RAS on AD, namely angiotensin II receptor blockers (ARBs) and angiotensin converting enzyme inhibitors (ACEIs), is highlighted and data supporting this hope will be presented, from in vitro and in vivo pre-clinical to clinical studies.
Collapse
|
19
|
Yang M, Sun D, Wang Y, Yan M, Zheng J, Ren J. Cognitive Impairment in Heart Failure: Landscape, Challenges, and Future Directions. Front Cardiovasc Med 2022; 8:831734. [PMID: 35198608 PMCID: PMC8858826 DOI: 10.3389/fcvm.2021.831734] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 12/30/2021] [Indexed: 12/20/2022] Open
Abstract
Heart failure (HF) is a major global healthcare problem accounting for substantial deterioration of prognosis. As a complex clinical syndrome, HF often coexists with multi-comorbidities of which cognitive impairment (CI) is particularly important. CI is increasing in prevalence among patients with HF and is present in around 40%, even up to 60%, of elderly patients with HF. As a potent and independent prognostic factor, CI significantly increases the hospitalization and mortality and decreases quality of life in patients with HF. There has been a growing awareness of the complex bidirectional interaction between HF and CI as it shares a number of common pathophysiological pathways including reduced cerebral blood flow, inflammation, and neurohumoral activations. Research that focus on the precise mechanism for CI in HF is still ever insufficient. As the tremendous adverse consequences of CI in HF, effective early diagnosis of CI in HF and interventions for these patients may halt disease progression and improve prognosis. The current clinical guidelines in HF have begun to emphasize the importance of CI. However, nearly half of CI in HF is underdiagnosed, and few recommendations are available to guide clinicians about how to approach CI in patients with HF. This review aims to synthesize knowledge about the link between HF and cognitive dysfunction, issues pertaining to screening, diagnosis and management of CI in patients with HF, and emerging therapies for prevention. Based on data from current studies, critical gaps in knowledge of CI in HF are identified, and future research directions to guide the field forward are proposed.
Collapse
Affiliation(s)
- Mengxi Yang
- Heart Failure Center, China-Japan Friendship Hospital, Beijing, China
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Di Sun
- Heart Failure Center, China-Japan Friendship Hospital, Beijing, China
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Yu Wang
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
| | - Mengwen Yan
- Heart Failure Center, China-Japan Friendship Hospital, Beijing, China
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Jingang Zheng
- Heart Failure Center, China-Japan Friendship Hospital, Beijing, China
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Jingyi Ren
- Heart Failure Center, China-Japan Friendship Hospital, Beijing, China
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
- Vascular Health Research Center of Peking University Health Science Center, Beijing, China
- *Correspondence: Jingyi Ren
| |
Collapse
|
20
|
Herrera P, Cauchi RJ. ACE and ACE2: insights from Drosophila and implications for COVID-19. Heliyon 2021; 7:e08555. [PMID: 34901515 PMCID: PMC8648576 DOI: 10.1016/j.heliyon.2021.e08555] [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: 08/25/2021] [Revised: 10/21/2021] [Accepted: 12/02/2021] [Indexed: 12/27/2022] Open
Abstract
Angiotensin-converting enzyme (ACE) and its homologue ACE2 are key regulators of the renin-angiotensin system and thereby cardiovascular function through their zinc-metallopeptidase activity on vasoactive peptides. ACE2 also serves as the receptor for the cellular entry of various coronaviruses including the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which is responsible for the coronavirus disease 2019 (COVID-19). The unprecedented scale of the COVID-19 pandemic has spurred the use of mammalian models to investigate the SARS-ACE2 relationship and knowledge gained from such research has accelerated development of vaccines and therapeutics. Recent studies have just started to underscore the utility of the fruit fly Drosophila melanogaster as a model system to study virus-host interactions and pathogenicity. Notably, the remarkable existence of catalytically functional ACE and ACE2 orthologues in Drosophila, discovered more than two decades ago, provides a unique opportunity for further developing this model organism to better understand COVID-19 in addition to identifying coronavirus preventative and therapeutic interventions targeting ACE2. Here, we review the studies that revealed crucial insights on the biochemistry and physiology of Ance and Acer, two out of the six Drosophila ACE family members with the greatest homology to human ACE and ACE2. We highlight shared in vivo functions outside of the renin-angiotensin system, which is not conserved in flies. Importantly, we identify knowledge gaps that can be filled by further research and outline ways that can raise Drosophila to a powerful model system to combat SARS-CoV-2 and its threatening vaccine-evading variants.
Collapse
Affiliation(s)
- Paul Herrera
- Centre for Molecular Medicine and Biobanking, Biomedical Sciences Building, University of Malta, Msida, Malta
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| | - Ruben J. Cauchi
- Centre for Molecular Medicine and Biobanking, Biomedical Sciences Building, University of Malta, Msida, Malta
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
- Corresponding author.
| |
Collapse
|
21
|
Zarfeshani A, Carroll KR, Volpe BT, Diamond B. Cognitive Impairment in SLE: Mechanisms and Therapeutic Approaches. Curr Rheumatol Rep 2021; 23:25. [PMID: 33782842 PMCID: PMC11207197 DOI: 10.1007/s11926-021-00992-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2021] [Indexed: 02/06/2023]
Abstract
A wide range of patients with systemic lupus erythematosus (SLE) suffer from cognitive dysfunction (CD) which severely impacts their quality of life. However, CD remains underdiagnosed and poorly understood. Here, we discuss current findings in patients and in animal models. Strong evidence suggests that CD pathogenesis involves known mechanisms of tissue injury in SLE. These mechanisms recruit brain resident cells, in particular microglia, into the pathological process. While systemic immune activation is critical to central nervous system injury, the current focus of therapy is the microglial cell and not the systemic immune perturbation. Further studies are critical to examine additional potential therapeutic targets and more specific treatments based on the cause and progress of the disease.
Collapse
Affiliation(s)
- Aida Zarfeshani
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Kaitlin R Carroll
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Bruce T Volpe
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Betty Diamond
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA.
| |
Collapse
|
22
|
Hernandez AR, Banerjee A, Carter CS, Buford TW. Angiotensin (1-7) Expressing Probiotic as a Potential Treatment for Dementia. FRONTIERS IN AGING 2021; 2:629164. [PMID: 34901930 PMCID: PMC8663799 DOI: 10.3389/fragi.2021.629164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 03/05/2021] [Indexed: 12/12/2022]
Abstract
Increasing life expectancies are unfortunately accompanied by increased prevalence of Alzheimer's disease (AD). Regrettably, there are no current therapeutic options capable of preventing or treating AD. We review here data indicating that AD is accompanied by gut dysbiosis and impaired renin angiotensin system (RAS) function. Therefore, we propose the potential utility of an intervention targeting both the gut microbiome and RAS as both are heavily involved in proper CNS function. One potential approach which our group is currently exploring is the use of genetically-modified probiotics (GMPs) to deliver therapeutic compounds. In this review, we specifically highlight the potential utility of utilizing a GMP to deliver Angiotensin (1-7), a beneficial component of the renin-angiotensin system with relevant functions in circulation as well as locally in the gut and brain.
Collapse
Affiliation(s)
- Abbi R. Hernandez
- Division of Gerontology, Geriatrics, and Palliative Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- UAB Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Anisha Banerjee
- Division of Gerontology, Geriatrics, and Palliative Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Christy S. Carter
- Division of Gerontology, Geriatrics, and Palliative Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- UAB Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Integrative Center for Aging Research, University of Alabama at Birmingham, Birmingham, AL, United States
- Nathan Shock Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Thomas W. Buford
- Division of Gerontology, Geriatrics, and Palliative Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- UAB Center for Exercise Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Integrative Center for Aging Research, University of Alabama at Birmingham, Birmingham, AL, United States
- Nathan Shock Center, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
23
|
Ouk M, Wu CY, Rabin JS, Jackson A, Edwards JD, Ramirez J, Masellis M, Swartz RH, Herrmann N, Lanctôt KL, Black SE, Swardfager W. The use of angiotensin-converting enzyme inhibitors vs. angiotensin receptor blockers and cognitive decline in Alzheimer's disease: the importance of blood-brain barrier penetration and APOE ε4 carrier status. ALZHEIMERS RESEARCH & THERAPY 2021; 13:43. [PMID: 33573702 PMCID: PMC7876820 DOI: 10.1186/s13195-021-00778-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 01/25/2021] [Indexed: 12/15/2022]
Abstract
Background The antihypertensive angiotensin receptor blockers (ARBs) and angiotensin-converting enzyme inhibitors (ACE-Is) have similar indications and mechanisms of action, but prior work suggests divergence in their effects on cognition. Methods Participants in the National Alzheimer’s Coordinating Center database with a clinical diagnosis of dementia due to Alzheimer’s disease (AD) using an ACE-I or an ARB at any visit were selected. The primary outcome was delayed recall memory on the Wechsler Memory Scale Revised – Logical Memory IIA. Other cognitive domains were explored, including attention and psychomotor processing speed (Trail Making Test [TMT]-A and Digit Symbol Substitution Test [DSST]), executive function (TMT-B), and language and semantic verbal fluency (Animal Naming, Vegetable Naming, and Boston Naming Tests). Random slopes mixed-effects models with inverse probability of treatment weighting were used, yielding rate ratios (RR) or regression coefficients (B), as appropriate to the distribution of the data. Apolipoprotein (APOE) ε4 status and blood-brain barrier (BBB) penetrance were investigated as effect modifiers. Results Among 1689 participants with AD, ARB use (n = 578) was associated with 9.4% slower decline in delayed recall performance over a mean follow-up of 2.28 years compared with ACE-I use (n = 1111) [RR = 1.094, p = 0.0327]; specifically, users of BBB-crossing ARBs (RR = 1.25, p = 0.002), BBB-crossing ACE-Is (RR = 1.16, p = 0.010), and non-BBB-crossing ARBs (RR = 1.20, p = 0.005) had better delayed recall performance over time compared with non-BBB-crossing ACE-I users. An interaction with APOE ε4 status (drug × APOE × time RR = 1.196, p = 0.033) emerged; ARBs were associated with better delayed recall scores over time than ACE-Is in non-carriers (RR = 1.200, p = 0.003), but not in carriers (RR = 1.003, p = 0.957). ARB use was also associated with better performance over time on the TMT-A (B = 2.023 s, p = 0.0004) and the DSST (B = 0.573 symbols, p = 0.0485), and these differences were significant among APOE ε4 non-carriers (B = 4.066 s, p = 0.0004; and B = 0.982 symbols, p = 0.0230; respectively). Some differences were seen also in language and verbal fluency among APOE ε4 non-carriers. Conclusions Among APOE ε4 non-carriers with AD, ARB use was associated with greater preservation of memory and attention/psychomotor processing speed, particularly compared to ACE-Is that do not cross the blood-brain-barrier. Supplementary Information The online version contains supplementary material available at 10.1186/s13195-021-00778-8.
Collapse
Affiliation(s)
- Michael Ouk
- Department of Pharmacology & Toxicology Room 4207, University of Toronto, Medical Sciences Building 1 King's College Circle, Toronto, ON, M5S 1A8, Canada.,Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Che-Yuan Wu
- Department of Pharmacology & Toxicology Room 4207, University of Toronto, Medical Sciences Building 1 King's College Circle, Toronto, ON, M5S 1A8, Canada.,Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Jennifer S Rabin
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada.,Harquail Centre for Neuromodulation, Sunnybrook Research Institute, Toronto, M4N 3M5, Canada.,Department of Medicine (Neurology), Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada.,Rehabilitation Sciences Institute, University of Toronto, Toronto, ON, Canada
| | - Aaron Jackson
- Department of Pharmacology & Toxicology Room 4207, University of Toronto, Medical Sciences Building 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
| | - Jodi D Edwards
- University of Ottawa Heart Institute, University of Ottawa, Ottawa, ON, Canada.,School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON, Canada.,ICES, Ottawa, ON, Canada
| | - Joel Ramirez
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada.,Canadian Partnership for Stroke Recovery, Toronto, ON, Canada
| | - Mario Masellis
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada.,Department of Medicine (Neurology), Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Richard H Swartz
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada.,Department of Medicine (Neurology), Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada.,Canadian Partnership for Stroke Recovery, Toronto, ON, Canada
| | - Nathan Herrmann
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada.,Department of Psychiatry, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Krista L Lanctôt
- Department of Pharmacology & Toxicology Room 4207, University of Toronto, Medical Sciences Building 1 King's College Circle, Toronto, ON, M5S 1A8, Canada.,Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada.,Department of Psychiatry, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada.,KITE UHN Toronto Rehabilitation Institute, Toronto, ON, Canada
| | - Sandra E Black
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada.,School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON, Canada.,Canadian Partnership for Stroke Recovery, Toronto, ON, Canada.,KITE UHN Toronto Rehabilitation Institute, Toronto, ON, Canada
| | - Walter Swardfager
- Department of Pharmacology & Toxicology Room 4207, University of Toronto, Medical Sciences Building 1 King's College Circle, Toronto, ON, M5S 1A8, Canada. .,Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada. .,Canadian Partnership for Stroke Recovery, Toronto, ON, Canada. .,KITE UHN Toronto Rehabilitation Institute, Toronto, ON, Canada.
| |
Collapse
|
24
|
Angiotensin Converting Enzyme Inhibitors and Angiotensin Receptor Blockers Rescue Memory Defects in Drosophila-Expressing Alzheimer's Disease-Related Transgenes Independently of the Canonical Renin Angiotensin System. eNeuro 2020; 7:ENEURO.0235-20.2020. [PMID: 33060184 PMCID: PMC7768280 DOI: 10.1523/eneuro.0235-20.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 09/29/2020] [Accepted: 10/01/2020] [Indexed: 01/09/2023] Open
Abstract
Alzheimer's disease (AD) is a degenerative disorder that causes progressive memory and cognitive decline. Recently, studies have reported that inhibitors of the mammalian renin angiotensin system (RAS) result in a significant reduction in the incidence and progression of AD by unknown mechanisms. Here, we used a genetic and pharmacological approach to evaluate the beneficial effects of angiotensin converting enzyme inhibitors (ACE-Is) and angiotensin receptor blockers (ARBs) in Drosophila expressing AD-related transgenes. Importantly, while ACE orthologs have been identified in Drosophila, other RAS components are not conserved. We show that captopril, an ACE-I, and losartan, an ARB, can suppress a rough eye phenotype and brain cell death in flies expressing a mutant human C99 transgene. Captopril also significantly rescues memory defects in these flies. Similarly, both drugs reduce cell death in Drosophila expressing human Aβ42 and losartan significantly rescues memory deficits. However, neither drug affects production, accumulation or clearance of Aβ42 Importantly, neither drug rescued brain cell death in Drosophila expressing human Tau, suggesting that RAS inhibitors specifically target the amyloid pathway. Of note, we also observed reduced cell death and a complete rescue of memory deficits when we crossed a null mutation in Drosophila Acer into each transgenic line demonstrating that the target of captopril in Drosophila is Acer. Together, these studies demonstrate that captopril and losartan are able to modulate AD related phenotypes in the absence of the canonical RAS pathway and suggest that both drugs have additional targets that can be identified in Drosophila.
Collapse
|
25
|
Lee H, Kim E. Repositioning medication for cardiovascular and cerebrovascular disease to delay the onset and prevent progression of Alzheimer's disease. Arch Pharm Res 2020; 43:932-960. [PMID: 32909178 DOI: 10.1007/s12272-020-01268-5] [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: 06/28/2020] [Accepted: 08/31/2020] [Indexed: 02/08/2023]
Abstract
Alzheimer's disease (AD) is a complex, progressive, neurodegenerative disorder. As with other common chronic diseases, multiple risk factors contribute to the onset and progression of AD. Many researchers have evaluated the epidemiologic and pathophysiological association between AD, cardiovascular diseases (CVDs), and cerebrovascular diseases (CBVDs), including commonly reported risk factors such as diabetes, hypertension, and dyslipidemia. Relevant therapies of CVDs/CBVDs for the attenuation of AD have also been empirically investigated. Considering the challenges of new drug development, in terms of cost and time, multifactorial approaches such as therapeutic repositioning of CVD/CBVD medication should be explored to delay the onset and progression of AD. Thus, in this review, we discuss our current understanding of the association between cardiovascular risk factors and AD, as revealed by clinical and non-clinical studies, as well as the therapeutic implications of CVD/CBVD medication that may attenuate AD. Furthermore, we discuss future directions by evaluating ongoing trials in the field.
Collapse
Affiliation(s)
- Heeyoung Lee
- Department of Clinical Medicinal Sciences, Konyang University, 121 Daehakro, Nonsan, 32992, Republic of Korea
| | - EunYoung Kim
- Evidence-Based Research Laboratory, Division of Clinical Pharmacotherapy, College of Pharmacy, Chung-Ang University, Seoul, 156-756, Republic of Korea.
| |
Collapse
|
26
|
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: 31] [Impact Index Per Article: 6.2] [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
|
27
|
Singh PK, Chen ZL, Ghosh D, Strickland S, Norris EH. Increased plasma bradykinin level is associated with cognitive impairment in Alzheimer's patients. Neurobiol Dis 2020; 139:104833. [PMID: 32173555 PMCID: PMC7175647 DOI: 10.1016/j.nbd.2020.104833] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 02/28/2020] [Accepted: 03/11/2020] [Indexed: 12/16/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by the presence of proteinaceous brain deposits, brain atrophy, vascular dysfunction, and chronic inflammation. Along with cerebral inflammation, peripheral inflammation is also evident in many AD patients. Bradykinin, a proinflammatory plasma peptide, is also linked to AD pathology. For example, bradykinin infusion into the hippocampus causes learning and memory deficits in rats, and blockade of the bradykinin receptor lessens cognitive impairment in AD mouse models. Even though it has been hypothesized that plasma bradykinin could contribute to inflammation in AD, the level of plasma bradykinin and its association with beta-amyloid (Aβ) pathology in AD patients had not been explored. Here, we assessed plasma bradykinin levels in AD patients and age-matched non-demented (ND) control individuals. We found significantly elevated plasma bradykinin levels in AD patients compared to ND subjects. Additionally, changes in plasma bradykinin levels were more profound in many AD patients with severe cognitive impairment, suggesting that peripheral bradykinin could play a role in dementia most likely via inflammation. Bradykinin levels in the cerebrospinal fluid (CSF) were reduced in AD patients and exhibited an inverse correlation with the CSF Aβ40/Aβ42 ratio. We also report that bradykinin interacts with the fibrillar form of Aβ and co-localizes with Aβ plaques in the post-mortem human AD brain. These findings connect the peripheral inflammatory pathway to cerebral abnormalities and identify a novel mechanism of inflammatory pathology in AD.
Collapse
Affiliation(s)
- Pradeep K Singh
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY 10065, USA
| | - Zu-Lin Chen
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY 10065, USA
| | - Dhiman Ghosh
- Laboratory of Physical Chemistry, ETH Zürich, WolfgangPauli-Str. 10, 8093 Zürich, Switzerland
| | - Sidney Strickland
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY 10065, USA
| | - Erin H Norris
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
28
|
Wright JW, Harding JW. Contributions by the Brain Renin-Angiotensin System to Memory, Cognition, and Alzheimer's Disease. J Alzheimers Dis 2020; 67:469-480. [PMID: 30664507 DOI: 10.3233/jad-181035] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by progressive neuron losses in memory-associated brain structures that rob patients of their dignity and quality of life. Five drugs have been approved by the FDA to treat AD but none modify or significantly slow disease progression. New therapies are needed to delay the course of this disease with the ultimate goal of preventing neuron losses and preserving memory functioning. In this review we describe the renin-angiotensin II (AngII) system (RAS) with specific regard to its deleterious contributions to hypertension, facilitation of neuroinflammation and oxidative stress, reduced cerebral blood flow, tissue remodeling, and disruption of memory consolidation and retrieval. There is evidence that components of the RAS, AngIV and Ang(1-7), are positioned to counter such damaging influences and these systems are detailed with the goal of drawing attention to their importance as drug development targets. Ang(1-7) binds at the Mas receptor, while AngIV binds at the AT4 receptor subtype, and these receptor numbers are significantly decreased in AD patients, accompanied by declines in brain aminopeptidases A and N, enzymes essential for the synthesis of AngIV. Potent analogs may be useful to counter these changes and facilitate neuronal functioning and reduce apoptosis in memory associated brain structures of AD patients.
Collapse
Affiliation(s)
- John W Wright
- Department of Psychology, Washington State University, Pullman, WA, USA.,Department of Integrative Physiology and Neuroscience, and Program in Biotechnology, Washington State University, Pullman, WA, USA.,M3 Biotechnology, Inc., Seattle, WA, USA
| | - Joseph W Harding
- Department of Psychology, Washington State University, Pullman, WA, USA.,Department of Integrative Physiology and Neuroscience, and Program in Biotechnology, Washington State University, Pullman, WA, USA.,M3 Biotechnology, Inc., Seattle, WA, USA
| |
Collapse
|
29
|
Cappetta K, Lago L, Potter J, Phillipson L. Under-coding of dementia and other conditions indicates scope for improved patient management: A longitudinal retrospective study of dementia patients in Australia. Health Inf Manag 2020; 51:32-44. [PMID: 31971019 DOI: 10.1177/1833358319897928] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Under-coding of dementia during hospitalisation results in an inability to identify all patients with dementia using hospital administrative data. Clinical coding can be viewed as a proxy for management; therefore, under-coding indicates dementia was not considered in the patient's management. While under-coding of dementia is well established, there is sparse evidence on whether dementia is coded in subsequent hospitalisations among patients with a known diagnosis. OBJECTIVE (a) To describe patterns of dementia coding over 5 years after a first-coded (i.e. index) admission for dementia; (b) to identify factors associated with clinical coding of dementia; and (c) to identify patient subgroups at risk of not being coded to inform future interventions to improve hospital identification and management of dementia. METHOD Retrospective study of longitudinal hospital data from 1 July 2006 to 30 June 2015 for 7919 patients hospitalised during the 5 years' post-index admission for dementia in a regional local health district of New South Wales, Australia. RESULTS Dementia was coded in 63.9% of admissions in the 12 months following index admission for dementia; this decreased to 53.7% after 5 years. Patients were 20% more likely to have dementia actively managed when it co-occurred with delirium. Under-coding varied across conditions, with dementia more likely to be coded in admissions for falls and pneumonitis, and less likely for heart failure, pneumonia and urinary tract infection (UTI). CONCLUSION The frequency with which dementia was not coded highlights opportunities to improve identification and management of dementia through dementia-specific care, enhanced clinical protocols, and interventions focused around heart failure, pneumonia and UTI admissions.
Collapse
Affiliation(s)
| | | | - Jan Potter
- University of Wollongong, Australia.,Illawarra Shoalhaven Local Health District, Australia
| | | |
Collapse
|
30
|
Anastasio TJ. Exploring the Correlation between the Cognitive Benefits of Drug Combinations in a Clinical Database and the Efficacies of the Same Drug Combinations Predicted from a Computational Model. J Alzheimers Dis 2019; 70:287-302. [PMID: 31177222 PMCID: PMC6700640 DOI: 10.3233/jad-190144] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Identification of drug combinations that could be effective in Alzheimer’s disease treatment is made difficult by the sheer number of possible combinations. This analysis identifies as potentially therapeutic those drug combinations that rank highest when their efficacy is determined jointly from two independent data sources. Estimates of the efficacy of the same drug combinations were derived from a clinical dataset on cognitively impaired elderly participants and from pre-clinical data, in the form of a computational model of neuroinflammation. Linear regression was used to show that the two sets of estimates were correlated, and to rule out confounds. The ten highest ranking, jointly determined drug combinations most frequently consisted of COX2 inhibitors and aspirin, along with various antihypertensive medications. Ten combinations of from five to nine drugs, and the three-drug combination of a COX2 inhibitor, aspirin, and a calcium-channel blocker, are discussed as candidates for consideration in future pre-clinical and clinical studies.
Collapse
Affiliation(s)
- Thomas J Anastasio
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| |
Collapse
|
31
|
Quitterer U, AbdAlla S. Improvements of symptoms of Alzheimer`s disease by inhibition of the angiotensin system. Pharmacol Res 2019; 154:104230. [PMID: 30991105 DOI: 10.1016/j.phrs.2019.04.014] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 04/10/2019] [Accepted: 04/11/2019] [Indexed: 01/30/2023]
Abstract
With ageing of the global society, the frequency of ageing-related neurodegenerative diseases such as Alzheimer`s disease (AD) is on the rise worldwide. Currently, there is no cure for AD, and the four drugs approved for AD only have very small effects on AD symptoms. Consequently, there are enormous efforts worldwide to identify new targets for treatment of AD. Approaches that interfere with classical neuropathologic features of AD, such as extracellular senile plaques formed of aggregated amyloid-beta (Abeta), and intracellular neurofibrillary tangles of hyperphosphorylated tau have not been successful so far. In search for a treatment approach of AD, we found that inhibition of the angiotensin-converting enzyme (ACE) by a centrally acting ACE inhibitor retards symptoms of neurodegeneration, Abeta plaque formation and tau hyperphosphorylation in experimental models of AD. Our approach is currently being investigated in a clinical setting. Initial evidence with AD patients shows that a brain-penetrating ACE inhibitor counteracts the process of neurodegeneration and dementia. Moreover, centrally acting ACE inhibitors given in addition to the standard therapy, cholinesterase inhibition, can improve cognitive function of AD patients for several months. This is one of the most promising results for AD treatment since more than a decade.
Collapse
Affiliation(s)
- Ursula Quitterer
- Molecular Pharmacology, Department of Chemistry and Applied Biosciences, ETH Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland; Institute of Pharmacology and Toxicology, Department of Medicine, University of Zurich, Winterthurerstrasse 190, CH-8057, Zürich, Switzerland.
| | - Said AbdAlla
- Molecular Pharmacology, Department of Chemistry and Applied Biosciences, ETH Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| |
Collapse
|
32
|
Fouda AY, Fagan SC, Ergul A. Brain Vasculature and Cognition. Arterioscler Thromb Vasc Biol 2019; 39:593-602. [PMID: 30816798 PMCID: PMC6540805 DOI: 10.1161/atvbaha.118.311906] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 02/15/2019] [Indexed: 12/18/2022]
Abstract
There is a complex interaction between the brain and the cerebral vasculature to meet the metabolic demands of the brain for proper function. Preservation of cerebrovascular function and integrity has a central role in this sophisticated communication within the brain, and any derangements can have deleterious acute and chronic consequences. In almost all forms of cognitive impairment, from mild to Alzheimer disease, there are changes in cerebrovascular function and structure leading to decreased cerebral blood flow, which may initiate or worsen cognitive impairment. In this focused review, we discuss the contribution of 2 major vasoactive pathways to cerebrovascular dysfunction and cognitive impairment in an effort to identify early intervention strategies.
Collapse
Affiliation(s)
- Abdelrahman Y. Fouda
- Vascular Biology Center, Augusta University, GA
- Charlie Norwood VA Medical Center Augusta, GA
| | - Susan C. Fagan
- Program in Clinical and Experimental Therapeutics, University of Georgia College of Pharmacy, GA
- Charlie Norwood VA Medical Center Augusta, GA
| | - Adviye Ergul
- Ralph Johnson Veterans Administration Medical Center, Medical University of South Carolina, Charleston, SC
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC
| |
Collapse
|
33
|
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: 29] [Impact Index Per Article: 4.1] [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
|
34
|
Hu Z, Wang L, Ma S, Kirisci L, Feng Z, Xue Y, Klunk WE, Kamboh MI, Sweet RA, Becker J, Lv Q, Lopez OL, Xie XQ. Synergism of antihypertensives and cholinesterase inhibitors in Alzheimer's disease. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2018; 4:542-555. [PMID: 30386819 PMCID: PMC6205113 DOI: 10.1016/j.trci.2018.09.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION We investigated the effect of antihypertensive (aHTN) medications and cholinesterase inhibitors (ChEIs) on the cognitive decline in patients with Alzheimer's disease (AD) and analyzed synergism by chemogenomics systems pharmacology mapping. METHODS We compared the effect of aHTN drugs on Mini-Mental State Examination scores in 617 AD patients with hypertension, and studied the synergistic effects. RESULTS The combination of diuretics, calcium channel blockers, and renin-angiotensin-aldosterone system blockers showed slower cognitive decline compared with other aHTN groups (Δβ = +1.46, P < .0001). aHTN medications slow down cognitive decline in ChEI users (Δβ = +0.56, P = .006), but not in non-ChEI users (Δβ = -0.31, P = .53). DISCUSSION aHTN and ChEI drugs showed synergistic effects. A combination of diuretics, renin-angiotensin-aldosterone system blockers, and calcium channel blockers had the slowest cognitive decline. The chemogenomics systems pharmacology-identified molecular targets provide system pharmacology interpretation of the synergism of the drugs in clinics. The results suggest that improving vascular health is essential for AD treatment and provide a novel direction for AD drug development.
Collapse
Affiliation(s)
- Ziheng Hu
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy; NIDA National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Lirong Wang
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy; NIDA National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Shifan Ma
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy; NIDA National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Levent Kirisci
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy; NIDA National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Zhiwei Feng
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy; NIDA National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ying Xue
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy; NIDA National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Clinical Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - William E. Klunk
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - M. Ilyas Kamboh
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Robert A. Sweet
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - James Becker
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Qianzhou Lv
- Department of Clinical Pharmacy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Oscar L. Lopez
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xiang-Qun Xie
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy; NIDA National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
35
|
Nestor J, Arinuma Y, Huerta TS, Kowal C, Nasiri E, Kello N, Fujieda Y, Bialas A, Hammond T, Sriram U, Stevens B, Huerta PT, Volpe BT, Diamond B. Lupus antibodies induce behavioral changes mediated by microglia and blocked by ACE inhibitors. J Exp Med 2018; 215:2554-2566. [PMID: 30185634 PMCID: PMC6170183 DOI: 10.1084/jem.20180776] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 07/06/2018] [Accepted: 08/02/2018] [Indexed: 02/03/2023] Open
Abstract
Cognitive impairment occurs in 40-90% of patients with systemic lupus erythematosus (SLE), which is characterized by autoantibodies to nuclear antigens, especially DNA. We discovered that a subset of anti-DNA antibodies, termed DNRAbs, cross reacts with the N-methyl-d-aspartate receptor (NMDAR) and enhances NMDAR signaling. In patients, DNRAb presence associates with spatial memory impairment. In a mouse model, DNRAb-mediated brain pathology proceeds through an acute phase of excitotoxic neuron loss, followed by persistent alteration in neuronal integrity and spatial memory impairment. The latter pathology becomes evident only after DNRAbs are no longer detectable in the brain. Here we investigate the mechanism of long-term neuronal dysfunction mediated by transient exposure to antibody. We show that activated microglia and C1q are critical mediators of neuronal damage. We further show that centrally acting inhibitors of angiotensin-converting enzyme (ACE) can prevent microglial activation and preserve neuronal function and cognitive performance. Thus, ACE inhibition represents a strong candidate for clinical trials aimed at mitigating cognitive dysfunction.
Collapse
Affiliation(s)
- Jacquelyn Nestor
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institute for Medical Research, Manhasset, NY
- Donald & Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY
| | - Yoshiyuki Arinuma
- Rheumatology and Infectious Diseases, Kitasato University School of Medicine, Kanagawa, Japan
| | - Tomás S Huerta
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institute for Medical Research, Manhasset, NY
- Donald & Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY
| | - Czeslawa Kowal
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institute for Medical Research, Manhasset, NY
| | - Elham Nasiri
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institute for Medical Research, Manhasset, NY
| | - Nina Kello
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institute for Medical Research, Manhasset, NY
| | - Yuichiro Fujieda
- Department of Rheumatology, Endocrinology and Nephrology Faculty of Medicine and Graduate School of Medicine Hokkaido University, Sapporo, Japan
| | - Alison Bialas
- Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Tim Hammond
- Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Uma Sriram
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Beth Stevens
- Kirby Neurobiology Center Boston Children's Hospital, Boston, MA
| | - Patricio T Huerta
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institute for Medical Research, Manhasset, NY
- Donald & Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY
| | - Bruce T Volpe
- Center for Biomedical Sciences, The Feinstein Institute for Medical Research, Manhasset, NY
| | - Betty Diamond
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, The Feinstein Institute for Medical Research, Manhasset, NY
| |
Collapse
|
36
|
Gebre AK, Altaye BM, Atey TM, Tuem KB, Berhe DF. Targeting Renin-Angiotensin System Against Alzheimer's Disease. Front Pharmacol 2018; 9:440. [PMID: 29760662 PMCID: PMC5937164 DOI: 10.3389/fphar.2018.00440] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 04/13/2018] [Indexed: 01/01/2023] Open
Abstract
Renin Angiotensin System (RAS) is a hormonal system that regulates blood pressure and fluid balance through a coordinated action of renal, cardiovascular, and central nervous systems. In addition to its hemodynamic regulatory role, RAS involves in many brain activities, including memory acquisition and consolidation. This review has summarized the involvement of RAS in the pathology of Alzheimer’s disease (AD), and the outcomes of treatment with RAS inhibitors. We have discussed the effect of brain RAS in the amyloid plaque (Aβ) deposition, oxidative stress, neuroinflammation, and vascular pathology which are directly and indirectly associated with AD. Angiotensin II (AngII) via AT1 receptor is reported to increase brain Aβ level via different mechanisms including increasing amyloid precursor protein (APP) mRNA, β-secretase activity, and presenilin expression. Similarly, it was associated with tau phosphorylation, and reactive oxygen species generation. However, these effects are counterbalanced by Ang II mediated AT2 signaling. The protective effect observed with angiotensin receptor blockers (ARBs) and angiotensin converting enzyme inhibitors (ACEIs) could be as the result of inhibition of Ang II signaling. ARBs also offer additional benefit by shifting the effect of Ang II toward AT2 receptor. To conclude, targeting RAS in the brain may benefit patients with AD though it still requires further in depth understanding.
Collapse
Affiliation(s)
- Abadi Kahsu Gebre
- Department of Pharmacology and Toxicology, School of Pharmacy, College of Health Sciences, Mekelle University, Mekelle, Ethiopia
| | - Birhanetensay Masresha Altaye
- Department of Pharmacology and Toxicology, School of Pharmacy, College of Health Sciences, Mekelle University, Mekelle, Ethiopia
| | - Tesfay Mehari Atey
- Clinical Pharmacy Unit, School of Pharmacy, College of Health Sciences, Mekelle University, Mekelle, Ethiopia
| | - Kald Beshir Tuem
- Department of Pharmacology and Toxicology, School of Pharmacy, College of Health Sciences, Mekelle University, Mekelle, Ethiopia
| | - Derbew Fikadu Berhe
- Department of Pharmacology and Toxicology, School of Pharmacy, College of Health Sciences, Mekelle University, Mekelle, Ethiopia
| |
Collapse
|
37
|
Etchegoyen M, Nobile MH, Baez F, Posesorski B, González J, Lago N, Milei J, Otero-Losada M. Metabolic Syndrome and Neuroprotection. Front Neurosci 2018; 12:196. [PMID: 29731703 PMCID: PMC5919958 DOI: 10.3389/fnins.2018.00196] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 03/12/2018] [Indexed: 12/17/2022] Open
Abstract
Introduction: Over the years the prevalence of metabolic syndrome (MetS) has drastically increased in developing countries as a major byproduct of industrialization. Many factors, such as the consumption of high-calorie diets and a sedentary lifestyle, bolster the spread of this disorder. Undoubtedly, the massive and still increasing incidence of MetS places this epidemic as an important public health issue. Hereon we revisit another outlook of MetS beyond its classical association with cardiovascular disease (CVD) and Diabetes Mellitus Type 2 (DM2), for MetS also poses a risk factor for the nervous tissue and threatens neuronal function. First, we revise a few essential concepts of MetS pathophysiology. Second, we explore some neuroprotective approaches in MetS pertaining brain hypoxia. The articles chosen for this review range from the years 1989 until 2017; the selection criteria was based on those providing data and exploratory information on MetS as well as those that studied innovative therapeutic approaches. Pathophysiology: The characteristically impaired metabolic pathways of MetS lead to hyperglycemia, insulin resistance (IR), inflammation, and hypoxia, all closely associated with an overall pro-oxidative status. Oxidative stress is well-known to cause the wreckage of cellular structures and tissue architecture. Alteration of the redox homeostasis and oxidative stress alter the macromolecular array of DNA, lipids, and proteins, in turn disrupting the biochemical pathways necessary for normal cell function. Neuroprotection: Different neuroprotective strategies are discussed involving lifestyle changes, medication aimed to mitigate MetS cardinal symptoms, and treatments targeted toward reducing oxidative stress. It is well-known that the routine practice of physical exercise, aerobic activity in particular, and a complete and well-balanced nutrition are key factors to prevent MetS. Nevertheless, pharmacological control of MetS as a whole and pertaining hypertension, dyslipidemia, and endothelial injury contribute to neuronal health improvement. Conclusion: The development of MetS has risen as a risk factor for neurological disorders. The therapeutic strategies include multidisciplinary approaches directed to address different pathological pathways all in concert.
Collapse
Affiliation(s)
- Melisa Etchegoyen
- Institute of Cardiological Research, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Mariana H Nobile
- Institute of Cardiological Research, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Francisco Baez
- Institute of Cardiological Research, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Barbara Posesorski
- Institute of Cardiological Research, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Julian González
- Institute of Cardiological Research, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Néstor Lago
- Institute of Cardiovascular Pathophysiology, School of Medicine, University of Buenos Aires, UBA-CONICET, Buenos Aires, Argentina
| | - José Milei
- Institute of Cardiological Research, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Matilde Otero-Losada
- Institute of Cardiological Research, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
38
|
Regenold WT, Blumenthal JB, Loreck DJ, Mordecai KL, Scarinzi G, Doddi SR, Adler L. Elevated Plasma Aβ42 in Cognitively Impaired Individuals Taking ACE Inhibitor Antihypertensives. Am J Alzheimers Dis Other Demen 2017; 32:347-352. [PMID: 28449585 PMCID: PMC10852825 DOI: 10.1177/1533317517707288] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND/RATIONALE Accumulating evidence suggests that the use of angiotensin-converting enzyme inhibitor (ACE-I) medication protects against cognitive decline in the elderly patients. We investigated whether ACE-I use was associated with higher plasma levels of amyloid-β (Aβ), possibly indicating improved Aβ clearance from brain to blood. METHODS We measured and compared plasma concentrations of Aβ42, Aβ40, and creatinine in cognitively impaired individuals with amnestic mild cognitive impairment, probable Alzheimer's disease (AD) dementia, and mixed probable AD/vascular dementia. RESULTS Plasma Aβ42 levels and Aβ42/Aβ40 ratios of participants taking ACE-Is (n = 11) significantly exceeded ( t = 3.1, df = 19, P = .006; U = 24, P = .029, respectively) those not taking ACE-Is (n = 10). CONCLUSIONS This study is the first to show an association between ACE-I use and increased plasma Aβ42 level and Aβ42/Aβ40 ratio in cognitively impaired individuals. Future investigations should assess whether a possible ACE-I-induced increase in plasma Aβ42 indicates improved Aβ42 clearance from brain that contributes to protection from cognitive decline.
Collapse
Affiliation(s)
- William T. Regenold
- Department of Psychiatry (Geriatric), University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jacob B. Blumenthal
- GRECC, Baltimore VA Medical Center, Baltimore, MD, USA
- Division of Gerontology and Geriatric Medicine/Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - David J. Loreck
- Department of Psychiatry (Geriatric), University of Maryland School of Medicine, Baltimore, MD, USA
- VA Maryland Health Care System, Baltimore, MD, USA
| | | | | | - Seshagiri R. Doddi
- Department of Psychiatry (Geriatric), University of Maryland School of Medicine, Baltimore, MD, USA
| | - Lawrence Adler
- Department of Psychiatry (Geriatric), University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
39
|
Li Y, Zhang Z, Deng L, Bai F, Shi Y, Yu H, Xie C, Yuan Y, Zhang Z. Genetic variation in angiotensin converting-enzyme affects the white matter integrity and cognitive function of amnestic mild cognitive impairment patients. J Neurol Sci 2017; 380:177-181. [PMID: 28870562 DOI: 10.1016/j.jns.2017.06.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Revised: 05/23/2017] [Accepted: 06/15/2017] [Indexed: 01/06/2023]
Abstract
Angiotensin-converting enzyme (ACE) gene has been implicated in amnestic mild cognitive impairment (aMCI). Most human genetic studies have focused on ACE insertion (I)/deletion (D) polymorphism and yielded conflicting results. In this work, we evaluated the relationships between cognitive function, serum ACE level, brain white matter (WM) integrity, and ACE I/D polymorphism in 48 patients with aMCI and 36 well matched control subjects from south China. In aMCI patients, D allele frequency was higher (D/I ratio=0.51:0.49) than that of the control subjects (D/I ratio=0.43:0.57); however, the difference was not statistically significant (p>0.05). The D carriers in aMCI subjects performed significantly poorer on auditory-verbal learning test (AVLT) -delayed recall than the I homozygous group (p=0.035). These carriers had higher serum ACE level than the I homozygous carriers of aMCI (p=0.037). In the aMCI group, D carriers showed significantly lower fractional anisotropy (FA) values in the left middle frontal gyrus, left anterior cingulate, right gyrus parahippocampalis, right inferior parietal lobule, and bilateral anterior central gyrus than the I homozygotes carriers. However, no significant difference was observed in FA values between I homozygotes and D carriers in the control subjects. The ACE D allele in aMCI patients may increase the risk of cognitive impairment. A high serum ACE level possibly plays an important role in the incidence of aMCI.
Collapse
Affiliation(s)
- Yanzhi Li
- Geriatric Department of Affiliated ZhongDa Hospital, Medical School of Southeast University, Nanjing, Jiangsu 210009, China
| | - Zhengsheng Zhang
- Neurologic Department of Affiliated ZhongDa Hospital, Neuropsychiatric Institute and Medical School of Southeast University, Nanjing, Jiangsu 210009, China
| | - Linglong Deng
- School of Clinical Medicine, Southeast University, Nanjing 210009, China
| | - Feng Bai
- Neurologic Department of Affiliated ZhongDa Hospital, Neuropsychiatric Institute and Medical School of Southeast University, Nanjing, Jiangsu 210009, China
| | - Yongmei Shi
- Neurologic Department of Affiliated ZhongDa Hospital, Neuropsychiatric Institute and Medical School of Southeast University, Nanjing, Jiangsu 210009, China
| | - Hui Yu
- School of Clinical Medicine, Southeast University, Nanjing 210009, China
| | - Chunming Xie
- Neurologic Department of Affiliated ZhongDa Hospital, Neuropsychiatric Institute and Medical School of Southeast University, Nanjing, Jiangsu 210009, China
| | - Yonggui Yuan
- Psychiatric Department of Affiliated ZhongDa Hospital, Medical School of Southeast University, Nanjing, Jiangsu 210009, China
| | - Zhijun Zhang
- Geriatric Department of Affiliated ZhongDa Hospital, Medical School of Southeast University, Nanjing, Jiangsu 210009, China.
| |
Collapse
|
40
|
Hernandorena I, Duron E, Vidal JS, Hanon O. Treatment options and considerations for hypertensive patients to prevent dementia. Expert Opin Pharmacother 2017; 18:989-1000. [PMID: 28532183 DOI: 10.1080/14656566.2017.1333599] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
INTRODUCTION Dementia is a worldwide health concern, which leads to loss of autonomy. To date no curative treatment is available so focus on modifiable risk factors is of particular interest. Hypertension, particularly midlife high blood pressure, has been associated with an increased risk for cognitive decline and dementia including vascular dementia (VAD) and Alzheimer disease (AD). In this context, antihypertensive treatments might have a preventive effect. The objective of this review was to examine the relationship between antihypertensive therapy and cognitive decline or dementia. Areas covered: A literature search was conducted using PUBMED and the COCHRANE LIBRARY for publications from 1990 onwards mentioning cognitive decline, AD, Vad, mixed dementia, vascular cognitive impairment, hypertension and antihypertensive therapy. Thirty-nine relevant publications including 20 longitudinal studies, 10 randomized-controlled trials and 9 meta-analyses were taken into account. Expert opinion: Most observational studies have suggested a potential preventive effect of antihypertensive therapies on cognitive decline and dementia, particularly calcium channel blockers and renin-angiotensin system blockers. Randomized clinical trials and meta-analyses provided more conflicting results potentially due to methodological issues. In conclusion, antihypertensive therapies may reduce cognitive decline and incidence of dementia. Further randomized clinical trials conducted in populations at higher risk of cognitive decline, with longer periods of follow-up and cognition as the primary outcome are still needed.
Collapse
Affiliation(s)
- Intza Hernandorena
- a Department of Geriatrics , APHP, Hôpital Broca , Paris , France.,b Sorbonne Paris Cité , University Descartes , Paris , France
| | - Emmanuelle Duron
- a Department of Geriatrics , APHP, Hôpital Broca , Paris , France.,b Sorbonne Paris Cité , University Descartes , Paris , France
| | - Jean-Sébastien Vidal
- a Department of Geriatrics , APHP, Hôpital Broca , Paris , France.,b Sorbonne Paris Cité , University Descartes , Paris , France
| | - Olivier Hanon
- a Department of Geriatrics , APHP, Hôpital Broca , Paris , France.,b Sorbonne Paris Cité , University Descartes , Paris , France
| |
Collapse
|
41
|
Santos CY, Snyder PJ, Wu WC, Zhang M, Echeverria A, Alber J. Pathophysiologic relationship between Alzheimer's disease, cerebrovascular disease, and cardiovascular risk: A review and synthesis. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2017; 7:69-87. [PMID: 28275702 PMCID: PMC5328683 DOI: 10.1016/j.dadm.2017.01.005] [Citation(s) in RCA: 243] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
As the population ages due to demographic trends and gains in life expectancy, the incidence and prevalence of dementia increases, and the need to understand the etiology and pathogenesis of dementia becomes ever more urgent. Alzheimer's disease (AD), the most common form of dementia, is a complex disease, the mechanisms of which are poorly understood. The more we learn about AD, the more questions are raised about our current conceptual models of disease. In the absence of a cure or the means by which to slow disease progress, it may be prudent to apply our current knowledge of the intersection between AD, cardiovascular disease, and cerebrovascular disease to foster efforts to delay or slow the onset of AD. This review discusses our current understanding of the epidemiology, genetics, and pathophysiology of AD, the intersection between AD and vascular causes of dementia, and proposes future directions for research and prevention.
Collapse
Affiliation(s)
- Cláudia Y. Santos
- Lifespan Clinical Research Center, Rhode Island Hospital, Providence, RI, USA
- Interdisciplinary Neuroscience Program, University of Rhode Island, Kingston, RI, USA
| | - Peter J. Snyder
- Lifespan Clinical Research Center, Rhode Island Hospital, Providence, RI, USA
- Interdisciplinary Neuroscience Program, University of Rhode Island, Kingston, RI, USA
- Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Wen-Chih Wu
- Division of Cardiology, Department of Medicine, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Mia Zhang
- Griffith University School of Medicine, Gold Coast, Queensland, Australia
| | - Ana Echeverria
- University of Puerto Rico School of Medicine, San Juan, Puerto Rico
| | - Jessica Alber
- Lifespan Clinical Research Center, Rhode Island Hospital, Providence, RI, USA
- Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI, USA
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School of Brown University, Providence, RI, USA
| |
Collapse
|
42
|
Rygiel K. Can angiotensin-converting enzyme inhibitors impact cognitive decline in early stages of Alzheimer's disease? An overview of research evidence in the elderly patient population. J Postgrad Med 2017; 62:242-248. [PMID: 27763482 PMCID: PMC5105210 DOI: 10.4103/0022-3859.188553] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease, in which an accumulation of toxic amyloid beta in the brain precedes the emergence of clinical symptoms. AD spectrum consists of presymptomatic, early symptomatic, and symptomatic phase of dementia. At present, no pharmacotherapy exists to modify or reverse a course of AD, and only symptomatic treatments are available. Many elderly patients, diagnosed with multiple medical conditions (such as cardiovascular diseases, Type 2 diabetes mellitus, and cerebrovascular diseases) are at increased risk of the development of mild cognitive impairment (MCI), AD, and vascular dementia. Studies have revealed reduced rates of cognitive decline, in elderly patients, who were treated with centrally active angiotensin-converting enzyme inhibitors (ACE-Is) (that have an ability to cross the blood–brain barrier). This article reviews recently published literature, focused on possible protective influence of the centrally active ACE-Is, in the elderly population, at risk for cognitive decline.
Collapse
Affiliation(s)
- K Rygiel
- Department of Family Practice, Medical University of Silesia (SUM), Katowice-Zabrze, 3 Maja St. 13/15, 41-800 Zabrze, Poland
| |
Collapse
|
43
|
Fekih-Mrissa N, Bedoui I, Sayeh A, Derbali H, Mrad M, Mrissa R, Nsiri B. Association between an angiotensin-converting enzyme gene polymorphism and Alzheimer's disease in a Tunisian population. Ann Gen Psychiatry 2017; 16:41. [PMID: 29176997 PMCID: PMC5693601 DOI: 10.1186/s12991-017-0164-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 11/08/2017] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND The angiotensin-converting enzyme gene (ACE) insertion/deletion (I/D or indel) polymorphism has long been linked to Alzheimer's disease (AD), but the interpretation of established data remains controversial. The aim of this study was to determine whether the angiotensin-converting enzyme is associated with the risk of Alzheimer's disease in Tunisian patients. METHODS We analyzed the genotype and allele frequency distribution of the ACE I/D gene polymorphism in 60 Tunisian AD patients and 120 healthy controls. RESULTS There is a significantly increased risk of AD in carriers of the D/D genotype (51.67% in patients vs. 31.67% in controls; p = .008, OR = 2.32). The D allele was also more frequently found in patients compared with controls (71.67% vs. 56.25%; p = .003, OR = 2.0). Moreover, as assessed by the Mini-Mental State Examination, patient D/D carriers were more frequently found to score in the severe category of dementia (65%) as compared to the moderate category (32%) or mild category (3%). CONCLUSIONS The D/D genotype and D allele of the ACE I/D polymorphism were associated with an increased risk in the development of AD in a Tunisian population. Furthermore, at the time of patient evaluation (average age 75 years), patients suffering with severe dementia were found predominantly in D/D carriers and, conversely, the D/D genotype and D allele were more frequently found in AD patients with severe dementia. These preliminary exploratory results should be confirmed in larger studies and further work is required to explore and interpret possible alternative findings in diverse populations.
Collapse
Affiliation(s)
- Najiba Fekih-Mrissa
- Laboratory of Molecular Biology, Department of Hematology, Military Hospital of Tunisia, Mont Fleury, 1008 Tunis, Tunisia
| | - Ines Bedoui
- Department of Neurology, Military Hospital of Tunisia, Montfleury, Tunis, 1008 Tunisia
| | - Aycha Sayeh
- Laboratory of Molecular Biology, Department of Hematology, Military Hospital of Tunisia, Mont Fleury, 1008 Tunis, Tunisia
| | - Hajer Derbali
- Department of Neurology, Military Hospital of Tunisia, Montfleury, Tunis, 1008 Tunisia
| | - Meriem Mrad
- Laboratory of Molecular Biology, Department of Hematology, Military Hospital of Tunisia, Mont Fleury, 1008 Tunis, Tunisia
| | - Ridha Mrissa
- Department of Neurology, Military Hospital of Tunisia, Montfleury, Tunis, 1008 Tunisia
| | - Brahim Nsiri
- Laboratory of Molecular Biology, Department of Hematology, Military Hospital of Tunisia, Mont Fleury, 1008 Tunis, Tunisia
| |
Collapse
|
44
|
Zhuang S, Wang X, Wang HF, Li J, Wang HY, Zhang HZ, Xing CM. Angiotensin converting enzyme serum activities: Relationship with Alzheimer’s disease. Brain Res 2016; 1650:196-202. [DOI: 10.1016/j.brainres.2016.09.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 07/24/2016] [Accepted: 09/04/2016] [Indexed: 12/19/2022]
|
45
|
Tan EC, Bell JS, Lu CY, Toh S. National Trends in Outpatient Antihypertensive Prescribing in People with Dementia in the United States. J Alzheimers Dis 2016; 54:1425-1435. [DOI: 10.3233/jad-160470] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Edwin C.K. Tan
- Centre for Medicine Use and Safety, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Parkville, Australia
- Aging Research Center, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | - J. Simon Bell
- Centre for Medicine Use and Safety, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Parkville, Australia
- Sansom Institute, University of South Australia, Adelaide, Australia
| | - Christine Y. Lu
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA
| | - Sengwee Toh
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA
| |
Collapse
|
46
|
Renin-angiotensin system as a potential therapeutic target in stroke and retinopathy: experimental and clinical evidence. Clin Sci (Lond) 2016; 130:221-38. [PMID: 26769658 DOI: 10.1042/cs20150350] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
As our knowledge expands, it is now clear that the renin-angiotensin (Ang) system (RAS) mediates functions other than regulating blood pressure (BP). The RAS plays a central role in the pathophysiology of different neurovascular unit disorders including stroke and retinopathy. Moreover, the beneficial actions of RAS modulation in brain and retina have been documented in experimental research, but not yet exploited clinically. The RAS is a complex system with distinct yet interconnected components. Understanding the different RAS components and their functions under brain and retinal pathological conditions is crucial to reap their benefits. The aim of the present review is to provide an experimental and clinical update on the role of RAS in the pathophysiology and treatment of stroke and retinopathy. Combining the evidence from both these disorders allows a unique opportunity to move both fields forward.
Collapse
|
47
|
Neasta J, Valmalle C, Coyne A, Carnazzi E, Subra G, Galleyrand J, Gagne D, M'Kadmi C, Bernad N, Bergé G, Cantel S, Marin P, Marie J, Banères J, Kemel M, Daugé V, Puget K, Martinez J. The novel nonapeptide acein targets angiotensin converting enzyme in the brain and induces dopamine release. Br J Pharmacol 2016; 173:1314-28. [PMID: 27027724 PMCID: PMC4940823 DOI: 10.1111/bph.13424] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2015] [Revised: 12/20/2016] [Accepted: 01/08/2016] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND AND PURPOSE Using an in-house bioinformatics programme, we identified and synthesized a novel nonapeptide, H-Pro-Pro-Thr-Thr-Thr-Lys-Phe-Ala-Ala-OH. Here, we have studied its biological activity, in vitro and in vivo, and have identified its target in the brain. EXPERIMENTAL APPROACH The affinity of the peptide was characterized using purified whole brain and striatal membranes from guinea pigs and rats . Its effect on behaviour in rats following intra-striatal injection of the peptide was investigated. A photoaffinity UV cross-linking approach combined with subsequent affinity purification of the ligand covalently bound to its receptor allowed identification of its target. KEY RESULTS The peptide bound with high affinity to a single class of binding sites, specifically localized in the striatum and substantia nigra of brains from guinea pigs and rats. When injected within the striatum of rats, the peptide stimulated in vitro and in vivo dopamine release and induced dopamine-like motor effects. We purified the target of the peptide, a ~151 kDa protein that was identified by MS/MS as angiotensin converting enzyme (ACE I). Therefore, we decided to name the peptide acein. CONCLUSION AND IMPLICATIONS The synthetic nonapeptide acein interacted with high affinity with brain membrane-bound ACE. This interaction occurs at a different site from the active site involved in the well-known peptidase activity, without modifying the peptidase activity. Acein, in vitro and in vivo, significantly increased stimulated release of dopamine from the brain. These results suggest a more important role for brain ACE than initially suspected.
Collapse
Affiliation(s)
- Jérémie Neasta
- Faculté de Pharmacie, Institut des Biomolécules Max Mousseron (IBMM) UMR 5247Université de Montpellier, CNRS, ENSCMMontpellierFrance
| | - Charlène Valmalle
- Faculté de Pharmacie, Institut des Biomolécules Max Mousseron (IBMM) UMR 5247Université de Montpellier, CNRS, ENSCMMontpellierFrance
| | - Anne‐Claire Coyne
- INSERM UMR 952, Physiopathologie des Maladies du Système Nerveux CentralParisFrance
| | - Eric Carnazzi
- Faculté de Pharmacie, Institut des Biomolécules Max Mousseron (IBMM) UMR 5247Université de Montpellier, CNRS, ENSCMMontpellierFrance
| | - Gilles Subra
- Faculté de Pharmacie, Institut des Biomolécules Max Mousseron (IBMM) UMR 5247Université de Montpellier, CNRS, ENSCMMontpellierFrance
| | - Jean‐Claude Galleyrand
- Faculté de Pharmacie, Institut des Biomolécules Max Mousseron (IBMM) UMR 5247Université de Montpellier, CNRS, ENSCMMontpellierFrance
| | - Didier Gagne
- Faculté de Pharmacie, Institut des Biomolécules Max Mousseron (IBMM) UMR 5247Université de Montpellier, CNRS, ENSCMMontpellierFrance
| | - Céline M'Kadmi
- Faculté de Pharmacie, Institut des Biomolécules Max Mousseron (IBMM) UMR 5247Université de Montpellier, CNRS, ENSCMMontpellierFrance
| | - Nicole Bernad
- Faculté de Pharmacie, Institut des Biomolécules Max Mousseron (IBMM) UMR 5247Université de Montpellier, CNRS, ENSCMMontpellierFrance
| | - Gilbert Bergé
- Faculté de Pharmacie, Institut des Biomolécules Max Mousseron (IBMM) UMR 5247Université de Montpellier, CNRS, ENSCMMontpellierFrance
| | - Sonia Cantel
- Faculté de Pharmacie, Institut des Biomolécules Max Mousseron (IBMM) UMR 5247Université de Montpellier, CNRS, ENSCMMontpellierFrance
| | - Philippe Marin
- Institut de Génomique Fonctionnelle, UMR5203, INSERM U661, Rue de la CardonilleUniversité de MontpellierMontpellierFrance
| | - Jacky Marie
- Faculté de Pharmacie, Institut des Biomolécules Max Mousseron (IBMM) UMR 5247Université de Montpellier, CNRS, ENSCMMontpellierFrance
| | - Jean‐Louis Banères
- Faculté de Pharmacie, Institut des Biomolécules Max Mousseron (IBMM) UMR 5247Université de Montpellier, CNRS, ENSCMMontpellierFrance
| | - Marie‐Lou Kemel
- CIRB, Collège de France, 11, Place Marcelin BerthelotParisFrance
| | - Valérie Daugé
- INSERM UMR 952, Physiopathologie des Maladies du Système Nerveux CentralParisFrance
| | - Karine Puget
- Faculté de Pharmacie, Institut des Biomolécules Max Mousseron (IBMM) UMR 5247Université de Montpellier, CNRS, ENSCMMontpellierFrance
| | - Jean Martinez
- Faculté de Pharmacie, Institut des Biomolécules Max Mousseron (IBMM) UMR 5247Université de Montpellier, CNRS, ENSCMMontpellierFrance
| |
Collapse
|
48
|
Dziedzic T, Pera J, Klimkowicz-Mrowiec A, Mroczko B, Slowik A. Biochemical and Radiological Markers of Alzheimer's Disease Progression. J Alzheimers Dis 2016; 50:623-44. [PMID: 26757184 DOI: 10.3233/ifs-150578] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative, inevitably progressive disease with a rate of cognitive, functional, and behavioral decline that varies highly from patient to patient. Although several clinical predictors of AD progression have been identified, to our mind in clinical practice there is a lack of a reliable biomarker that enables one to stratify the risk of deterioration. Identification of biomarkers that allow the monitoring of AD progression could change the way physicians and caregivers make treatment decisions. This review summarizes the results of studies on potential biochemical and radiological markers related to AD progression.
Collapse
Affiliation(s)
- Tomasz Dziedzic
- Department of Neurology, Jagiellonian University, Krakow, Poland
| | - Joanna Pera
- Department of Neurology, Jagiellonian University, Krakow, Poland
| | | | - Barbara Mroczko
- Department of Neurodegeneration Diagnostics, Medical University of Białystok, Poland.,Department of Biochemical Diagnostics, University Hospital, Białystok, Poland
| | - Agnieszka Slowik
- Department of Neurology, Jagiellonian University, Krakow, Poland
| |
Collapse
|
49
|
Inhibition of ACE Retards Tau Hyperphosphorylation and Signs of Neuronal Degeneration in Aged Rats Subjected to Chronic Mild Stress. BIOMED RESEARCH INTERNATIONAL 2015; 2015:917156. [PMID: 26697495 PMCID: PMC4677170 DOI: 10.1155/2015/917156] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 11/12/2015] [Indexed: 12/18/2022]
Abstract
With increasing life expectancy, Alzheimer's disease (AD) and other types of age-associated dementia are on the rise worldwide. Treatment approaches for dementia are insufficient and novel therapies are not readily available. In this context repurposing of established drugs appears attractive. A well-established class of cardiovascular drugs, which targets the angiotensin II system, is such a candidate, which currently undergoes a paradigm shift with regard to the potential benefit for treatment of neurodegenerative symptoms. In search for additional evidence, we subjected aged rats to chronic unpredictable mild stress, which is known to enhance the development of AD-related neuropathological features. We report here that four weeks of chronic mild stress induced a strong upregulation of the hippocampal angiotensin-converting enzyme (Ace) at gene expression and protein level. Concomitantly, tau protein hyperphosphorylation developed. Signs of neurodegeneration were detected by the significant downregulation of neuronal structure proteins such as microtubule-associated protein 2 (Map2) and synuclein-gamma (Sncg). Ace was involved in neurodegenerative symptoms because treatment with the brain-penetrating ACE inhibitor, captopril, retarded tau hyperphosphorylation and signs of neurodegeneration. Moreover, ACE inhibitor treatment could counteract glutamate neurotoxicity by preventing the downregulation of glutamate decarboxylase 2 (Gad2). Taken together, ACE inhibition targets neurodegeneration triggered by environmental stress.
Collapse
|
50
|
Ye R, Hu Y, Yao A, Yang Y, Shi Y, Jiang Y, Zhang J. Impact of renin-angiotensin system-targeting antihypertensive drugs on treatment of Alzheimer's disease: a meta-analysis. Int J Clin Pract 2015; 69:674-81. [PMID: 25721930 DOI: 10.1111/ijcp.12626] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
OBJECTIVE The impacts of renin-angiotensin system (RAS)-targeting antihypertensive drugs on Alzheimer's disease (AD) remain controversial. We performed this meta-analysis to evaluate the precise value of RAS-targeting antihypertensive drugs in terms of attenuating incidence of AD and slowing down cognitive decline in patients with AD. METHODS A systematic literature search was conducted using PubMed, Embase, ScienceDirect and CBM (China Biology Medicine Disc) before September 2014. Studies analysing incidence of AD and cognitive changes in AD patients with RAS-targeting antihypertensive drugs were identified. The principal outcome measures were hazard ratios (HRs) for incidence of AD and standardised mean difference (SMD) for cognitive changes in AD patients. Pooled data were calculated using fixed or random effects models according to the heterogeneity. RESULTS In total, 12 studies involving 896,410 participants met our inclusion criteria. RAS-targeting antihypertensive drugs were significantly associated with a reduced incidence rate of AD (HR 0.81, 95% CI 0.72-0.92, p = 0.001). In subgroup analysis, both angiotensin renin blockers and angiotensin converting enzyme inhibitors were shown to effectively decrease the incidence rate of AD. In the analysis of cognitive changes, a slower rate of cognitive decline was observed in AD patients with RAS-targeting antihypertensive drug (SMD 0.30, 95% CI 0.09-0.50, p = 0.004), when randomised trials and observational trials were combined. However, analysis of randomised trials alone did not show the same result (SMD 0.20, 95% CI -0.10 to 0.50, p = 0.182). CONCLUSIONS Renin-angiotensin system-targeting antihypertensive drugs may be a potential treatment for reducing the incidence and progression of AD. Further studies on RAS-targeting antihypertensive drugs, especially large randomised clinical trials, should be conducted in the future.
Collapse
Affiliation(s)
- R Ye
- Department of Neurology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Y Hu
- Department of Neurology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - A Yao
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Y Yang
- Department of Neurology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Y Shi
- Department of Neurology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Y Jiang
- Department of Neurology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - J Zhang
- Department of Neurology, Zhongnan Hospital, Wuhan University, Wuhan, China
| |
Collapse
|