1
|
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: 13.5] [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
|
2
|
Wu H, Sun Q, Yuan S, Wang J, Li F, Gao H, Chen X, Yang R, Xu J. AT1 Receptors: Their Actions from Hypertension to Cognitive Impairment. Cardiovasc Toxicol 2022; 22:311-325. [PMID: 35211833 PMCID: PMC8868040 DOI: 10.1007/s12012-022-09730-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 02/07/2022] [Indexed: 12/18/2022]
Abstract
Hypertension is one of the most prevalent cardiovascular disorders worldwide, affecting 1.13 billion people, or 14% of the global population. Hypertension is the single biggest risk factor for cerebrovascular dysfunction. According to the American Heart Association, high blood pressure (BP), especially in middle-aged individuals (~ 40 to 60 years old), is associated with an increased risk of dementia, later in life. Alzheimer’s disease and cerebrovascular disease are the two leading causes of dementia, accounting for around 80% of the total cases and usually combining mixed pathologies from both. Little is known regarding how hypertension affects cognitive function, so the impact of its treatment on cognitive impairment has been difficult to assess. The brain renin-angiotensin system (RAS) is essential for BP regulation and overactivity of this system has been established to precede the development and maintenance of hypertension. Angiotensin II (Ang-II), the main peptide within this system, induces vasoconstriction and impairs neuro-vascular coupling by acting on brain Ang-II type 1 receptors (AT1R). In this review, we systemically analyzed the association between RAS and biological mechanisms of cognitive impairment, from the perspective of AT1R located in the central nervous system. Additionally, the possible contribution of brain AT1R to global cognition decline in COVID-19 cases will be discussed as well.
Collapse
Affiliation(s)
- Hanxue Wu
- Department of Physiology and Pathophysiology, School of Medicine, Xi'an Jiaotong University, 76 West Yanta Road, Xi'an, 710061, China
| | - Qi Sun
- The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Shenglan Yuan
- Department of Physiology and Pathophysiology, School of Medicine, Xi'an Jiaotong University, 76 West Yanta Road, Xi'an, 710061, China
| | - Jiawei Wang
- Department of Physiology and Pathophysiology, School of Medicine, Xi'an Jiaotong University, 76 West Yanta Road, Xi'an, 710061, China
| | - Fanni Li
- The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Hongli Gao
- Department of Physiology and Pathophysiology, School of Medicine, Xi'an Jiaotong University, 76 West Yanta Road, Xi'an, 710061, China
| | - Xingjuan Chen
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Rui Yang
- Department of Physiology and Pathophysiology, School of Medicine, Xi'an Jiaotong University, 76 West Yanta Road, Xi'an, 710061, China
| | - Jiaxi Xu
- Department of Physiology and Pathophysiology, School of Medicine, Xi'an Jiaotong University, 76 West Yanta Road, Xi'an, 710061, China.
| |
Collapse
|
3
|
Hemorphins-From Discovery to Functions and Pharmacology. Molecules 2021; 26:molecules26133879. [PMID: 34201982 PMCID: PMC8270332 DOI: 10.3390/molecules26133879] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/18/2021] [Accepted: 06/21/2021] [Indexed: 01/31/2023] Open
Abstract
During the last three decades, a variety of different studies on bioactive peptides that are opioid receptor ligands, have been carried out, with regard to their isolation and identification, as well as their molecular functions in living organisms. Thus, in this review, we would like to summarize the present state-of-the art concerning hemorphins, methodological aspects of their identification, and their potential role as therapeutic agents. We have collected and discussed articles describing hemorphins, from their discovery up until now, thus presenting a very wide spectrum of their characteristic and applications. One of the major assets of the present paper is a combination of analytical and pharmacological aspects of peptides described by a team who participated in the initial research on hemorphins. This review is, in part, focused on the analysis of endogenous opioid peptides in biological samples using advanced techniques, description of the identification of synthetic/endogenous hemorphins, their involvement in pharmacology, learning, pain and other function. Finally, the part regarding hemorphin analogues and their synthesis, has been added.
Collapse
|
4
|
Ribeiro VT, de Souza LC, Simões E Silva AC. Renin-Angiotensin System and Alzheimer's Disease Pathophysiology: From the Potential Interactions to Therapeutic Perspectives. Protein Pept Lett 2020; 27:484-511. [PMID: 31886744 DOI: 10.2174/0929866527666191230103739] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 08/27/2019] [Accepted: 11/16/2019] [Indexed: 12/21/2022]
Abstract
New roles of the Renin-Angiotensin System (RAS), apart from fluid homeostasis and Blood Pressure (BP) regulation, are being progressively unveiled, since the discoveries of RAS alternative axes and local RAS in different tissues, including the brain. Brain RAS is reported to interact with pathophysiological mechanisms of many neurological and psychiatric diseases, including Alzheimer's Disease (AD). Even though AD is the most common cause of dementia worldwide, its pathophysiology is far from elucidated. Currently, no treatment can halt the disease course. Successive failures of amyloid-targeting drugs have challenged the amyloid hypothesis and increased the interest in the inflammatory and vascular aspects of AD. RAS compounds, both centrally and peripherally, potentially interact with neuroinflammation and cerebrovascular regulation. This narrative review discusses the AD pathophysiology and its possible interaction with RAS, looking forward to potential therapeutic approaches. RAS molecules affect BP, cerebral blood flow, neuroinflammation, and oxidative stress. Angiotensin (Ang) II, via angiotensin type 1 receptors may promote brain tissue damage, while Ang-(1-7) seems to elicit neuroprotection. Several studies dosed RAS molecules in AD patients' biological material, with heterogeneous results. The link between AD and clinical conditions related to classical RAS axis overactivation (hypertension, heart failure, and chronic kidney disease) supports the hypothesized role of this system in AD. Additionally, RAStargeting drugs as Angiotensin Converting Enzyme inhibitors (ACEis) and Angiotensin Receptor Blockers (ARBs) seem to exert beneficial effects on AD. Results of randomized controlled trials testing ACEi or ARBs in AD are awaited to elucidate whether AD-RAS interaction has implications on AD therapeutics.
Collapse
Affiliation(s)
- Victor Teatini Ribeiro
- Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Leonardo Cruz de Souza
- Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil.,Department of Internal Medicine, Service of Neurology, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Ana Cristina Simões E Silva
- Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| |
Collapse
|
5
|
Vear A, Gaspari T, Thompson P, Chai SY. Is There an Interplay Between the Functional Domains of IRAP? Front Cell Dev Biol 2020; 8:585237. [PMID: 33134302 PMCID: PMC7550531 DOI: 10.3389/fcell.2020.585237] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 09/08/2020] [Indexed: 01/16/2023] Open
Abstract
As a member of the M1 family of aminopeptidases, insulin regulated aminopeptidase (IRAP) is characterized by distinct binding motifs at the active site in the C-terminal domain that mediate the catalysis of peptide substrates. However, what makes IRAP unique in this family of enzymes is that it also possesses trafficking motifs at the N-terminal domain which regulate the movement of IRAP within different intracellular compartments. Research on the role of IRAP has focused predominantly on the C-terminus catalytic domain in different physiological and pathophysiological states ranging from pregnancy to memory loss. Many of these studies have utilized IRAP inhibitors, that bind competitively to the active site of IRAP, to explore the functional significance of its catalytic activity. However, it is unknown whether these inhibitors are able to access intracellular sites where IRAP is predominantly located in a basal state as the enzyme may need to be at the cell surface for the inhibitors to mediate their effects. This property of IRAP has often been overlooked. Interestingly, in some pathophysiological states, the distribution of IRAP is altered. This, together with the fact that IRAP possesses trafficking motifs, suggest the localization of IRAP may play an important role in defining its physiological or pathological functions and provide insights into the interplay between the two functional domains of the protein.
Collapse
Affiliation(s)
- Anika Vear
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Tracey Gaspari
- Department of Pharmacology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Philip Thompson
- Department of Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Siew Yeen Chai
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| |
Collapse
|
6
|
Royea J, Hamel E. Brain angiotensin II and angiotensin IV receptors as potential Alzheimer's disease therapeutic targets. GeroScience 2020; 42:1237-1256. [PMID: 32700176 DOI: 10.1007/s11357-020-00231-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 07/07/2020] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that is multifactorial in nature. Yet, despite being the most common form of dementia in the elderly, AD's primary cause remains unknown. As such, there is currently little to offer AD patients as the vast majority of recently tested therapies have either failed in well-controlled clinical trials or inadequately treat AD. Recently, emerging preclinical and clinical evidence has associated the brain renin angiotensin system (RAS) to AD pathology. Accordingly, various components of the brain RAS were shown to be altered in AD patients and mouse models, including the angiotensin II type 1 (AT1R), angiotensin IV receptor (AT4R), and Mas receptors. Collectively, the changes observed within the RAS have been proposed to contribute to many of the neuropathological hallmarks of AD, including the neuronal, cognitive, and vascular dysfunctions. Accumulating evidence has additionally identified antihypertensive medications targeting the RAS, particularly angiotensin receptor blockers (ARBs) and angiotensin-converting enzyme inhibitors (ACEIs), to delay AD onset and progression. In this review, we will discuss the emergence of the RAS's involvement in AD and highlight putative mechanisms of action underlying ARB's beneficial effects that may explain their ability to modify the risk of developing AD or AD progression. The RAS may provide novel molecular targets for recovering memory pathways, cerebrovascular function, and other pathological landmarks of AD.
Collapse
Affiliation(s)
- Jessika Royea
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, 3801 University Street, Montréal, QC, H3A 2B4, Canada
| | - Edith Hamel
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, 3801 University Street, Montréal, QC, H3A 2B4, Canada.
| |
Collapse
|
7
|
Visniauskas B, Simões PSR, Dalio FM, Naffah-Mazzacoratti MDG, Oliveira V, Tufik S, Chagas JR. Sleep deprivation changes thimet oligopeptidase (THOP1) expression and activity in rat brain. Heliyon 2019; 5:e02896. [PMID: 31828230 PMCID: PMC6889027 DOI: 10.1016/j.heliyon.2019.e02896] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 10/01/2019] [Accepted: 11/18/2019] [Indexed: 12/27/2022] Open
Abstract
The consequences of sleep deprivation on memory, cognition, nociception, stress, and endocrine function are related to the balance of neuropeptides, with peptidases being particularly essential. Thimet oligopeptidase (THOP1) is a metallopeptidase implicated in the metabolism of many sleep-related peptides, including angiotensin I, gonadotropin releasing hormone (GnRH), neurotensin, and opioid peptides. In the present study, we evaluated the effect of sleep deprivation and sleep recovery in male rats on THOP1 expression and specific activity in the central nervous system. In the striatum and hypothalamus, THOP1 activity decreased following sleep deprivation and a recovery period. Meanwhile, THOP1 activity and immunoexpression increased in the hippocampal dentate gyrus during the sleep recovery period. Changes in THOP1 expression after sleep deprivation and during sleep recovery can potentially alter the processing of neuropeptides. In particular, processing of opioid peptides may be related to the known increase in pain sensitivity in this model. These results suggest that THOP1 may be an important player in the effects of sleep deprivation.
Collapse
Affiliation(s)
- Bruna Visniauskas
- Department of Psychobiology, Universidade Federal de São Paulo, São Paulo, 04024-002, Brazil
| | - Priscila S R Simões
- Department of Neurology/Neurosurgery, Universidade Federal de São Paulo, São Paulo, 04039-032, Brazil
| | - Fernanda M Dalio
- Department of Biophysics, Universidade Federal de São Paulo, São Paulo, 04039-032, Brazil
| | | | - Vitor Oliveira
- Department of Biophysics, Universidade Federal de São Paulo, São Paulo, 04039-032, Brazil
| | - Sergio Tufik
- Department of Psychobiology, Universidade Federal de São Paulo, São Paulo, 04024-002, Brazil
| | - Jair R Chagas
- Department of Psychobiology, Universidade Federal de São Paulo, São Paulo, 04024-002, Brazil.,Department of Biophysics, Universidade Federal de São Paulo, São Paulo, 04039-032, Brazil
| |
Collapse
|
8
|
Royea J, Martinot P, Hamel E. Memory and cerebrovascular deficits recovered following angiotensin IV intervention in a mouse model of Alzheimer's disease. Neurobiol Dis 2019; 134:104644. [PMID: 31669735 DOI: 10.1016/j.nbd.2019.104644] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 10/01/2019] [Accepted: 10/16/2019] [Indexed: 10/25/2022] Open
Abstract
Angiotensin II type 1 receptor antagonists like losartan have been found to lower the incidence and progression to Alzheimer's disease (AD), as well as rescue cognitive and cerebrovascular deficits in AD mouse models. We previously found that co-administration of an angiotensin IV (AngIV) receptor (AT4R) antagonist prevented losartan's benefits, identifying AT4Rs as a possible target to counter AD pathogenesis. Therein, we investigated whether directly targeting AT4Rs could counter AD pathogenesis in a well-characterized mouse model of AD. Wild-type and human amyloid precursor protein (APP) transgenic (J20 line) mice (4.5 months old) received vehicle or AngIV (~1.3 nmol/day, 1 month) intracerebroventricularly via osmotic minipumps. AngIV restored short-term memory, spatial learning and memory in APP mice. AngIV normalized hippocampal AT4R levels, increased hippocampal subgranular zone cellular proliferation and dendritic arborization, and reduced oxidative stress. AngIV rescued whisker-evoked neurovascular coupling, endothelial- and smooth muscle cell-mediated cerebral vasodilatory responses, and cerebrovascular nitric oxide bioavailability. AngIV did not alter blood pressure, neuroinflammation or amyloid-β (Aβ) pathology. These preclinical findings identify AT4R as a promising target to counter Aβ-related cognitive and cerebrovascular deficits in AD.
Collapse
Affiliation(s)
- Jessika Royea
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada
| | - Pauline Martinot
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada
| | - Edith Hamel
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada.
| |
Collapse
|
9
|
Ho JK, Nation DA. Cognitive benefits of angiotensin IV and angiotensin-(1-7): A systematic review of experimental studies. Neurosci Biobehav Rev 2018; 92:209-225. [PMID: 29733881 PMCID: PMC8916541 DOI: 10.1016/j.neubiorev.2018.05.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 03/23/2018] [Accepted: 05/02/2018] [Indexed: 12/19/2022]
Abstract
OBJECTIVES To explore effects of the brain renin-angiotensin system (RAS) on cognition. DESIGN Systematic review of experimental (non-human) studies assessing cognitive effects of RAS peptides angiotensin-(3-8) [Ang IV] and angiotensin-(1-7) [Ang-(1-7)] and their receptors, the Ang IV receptor (AT4R) and the Mas receptor. RESULTS Of 450 articles identified, 32 met inclusion criteria. Seven of 11 studies of normal animals found Ang IV had beneficial effects on tests of passive or conditioned avoidance and object recognition. In models of cognitive deficit, eight of nine studies found Ang IV and its analogs (Nle1-Ang IV, dihexa, LVV-hemorphin-7) improved performance on spatial working memory and passive avoidance tasks. Two of three studies examining Ang-(1-7) found it benefited memory. Mas receptor removal was associated with reduced fear memory in one study. CONCLUSION Studies of cognitive impairment show salutary effects of acute administration of Ang IV and its analogs, as well as AT4R activation. Brain RAS peptides appear most effective administered intracerebroventricularly, close to the time of learning acquisition or retention testing. Ang-(1-7) shows anti-dementia qualities.
Collapse
Affiliation(s)
- Jean K Ho
- Department of Psychology, University of Southern California, Los Angeles, CA, USA.
| | - Daniel A Nation
- Department of Psychology, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
10
|
ATP6AP2 over-expression causes morphological alterations in the hippocampus and in hippocampus-related behaviour. Brain Struct Funct 2018; 223:2287-2302. [DOI: 10.1007/s00429-018-1633-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 02/18/2018] [Indexed: 01/07/2023]
|
11
|
Abstract
The classical renin-angiotensin system (RAS) in the body has been studied intensively in the last decades, since it is known that this system is involved in the regulation of blood pressure. Since nearly all members of the classical RAS have also been identified within the brain in the last decades and due to the existence of the blood-brain barrier, a RAS within the brain (bRAS) that is largely independent from the peripheral RAS has been postulated. All members of the angiotensin family as e.g., angiotensin II, angiotensin IV and angiotensin II (1-7) along with the respective receptors (e.g., angiotensin II receptor type 1 (AT1), angiotensin II receptor type 2 (AT2), angiotensin IV receptor (AT4), angiotensin II (1-7) receptor (Mas)) have been identified within the brain. Moreover, a receptor capable of binding renin and the renin precursor prorenin with high affinity has also been detected within the brain. This protein functions as a membrane receptor for (pro)renin and also represents a V-ATPase subunit and is therefore termed (P)RR or Atp6ap2, respectively. In this review we shed light on the (known as well as putative) roles and functions of Atp6ap2 in the brain under physiological and pathophysiological conditions.
Collapse
Affiliation(s)
- Alexander Bracke
- Institute of Anatomy and Cell Biology, Universitätsmedizin Greifswald, Greifswald, Germany
| | | |
Collapse
|
12
|
Singh Y, Gupta G, Shrivastava B, Dahiya R, Tiwari J, Ashwathanarayana M, Sharma RK, Agrawal M, Mishra A, Dua K. Calcitonin gene-related peptide (CGRP): A novel target for Alzheimer's disease. CNS Neurosci Ther 2017; 23:457-461. [PMID: 28417590 PMCID: PMC6492742 DOI: 10.1111/cns.12696] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 03/11/2017] [Accepted: 03/17/2017] [Indexed: 12/22/2022] Open
Abstract
Alzheimer's disease (AD) is leading cause of death among older characterized by neurofibrillary tangles, oxidative stress, progressive neuronal deficits, and increased levels of amyloid-β (Aβ) peptides. Cholinergic treatment could be the best suitable physiological therapy for AD. Calcitonin gene-related peptide (CGRP) is a thirty-seven-amino acid regulatory neuropeptide resulting from different merging of the CGRP gene, which also includes adrenomedullin, amylin, calcitonin, intermedin, and calcitonin receptor-stimulating peptide. It is a proof for a CGRP receptor within nucleus accumbens of brain that is different from either the CGRP1 or CGRP2 receptor in which it demonstrates similar high-affinity binding for salmon calcitonin, CGRP, and amylin, a possession which is not shared by any extra CGRP receptors. Binding of CGRP to its receptor increases activated cAMP-dependent pkA and PI3 kinase, resulting in N-terminal fragments that are shown to exert complex inhibitory as well facilitator actions on nAChRs. Fragments such as CGRP1-4, CGRP1-5, and CGRP1-6 rapidly as well as reversibly improve agonist sensitivity of nAChRs without straight stimulating those receptors and produce the Ca2+ -induced intracellular Ca2+ mobilization. Renin-angiotensin-aldosterone system (RAAS)-activated angiotensin-type (AT4) receptor is also beneficial in AD. It has been suggested that exogenous administration of CGRP inhibits infiltration of macrophages and expression of various inflammatory mediators such as NFkB, IL-1b, TNF-α, iNOS, matrix metalloproteinase (MMP)-9, and cell adhesion molecules like intercellular adhesion molecule (ICAM)-1 which attenuates consequence of inflammation in AD. Donepezil, a ChEI, inhibits acetylcholinesterase and produces angiogenesis and neurogenesis, in the dentate gyrus of the hippocampus of WT mice after donepezil administration. However, none of the results discovered in CGRP-knockout mice and WT mice exposed to practical denervation. Therefore, selective agonists of CGRP receptors may become the potential candidates for treatment of AD.
Collapse
Affiliation(s)
- Yogendra Singh
- School of PharmacyJaipur National UniversityJagatpuraJaipurIndia
| | - Gaurav Gupta
- School of PharmacyJaipur National UniversityJagatpuraJaipurIndia
- School of Medicine and Public HealthUniversity of NewcastleNewcastleNSWAustralia
| | | | - Rajiv Dahiya
- Laboratory of Peptide Research and DevelopmentSchool of Pharmacy, The University of the West IndiesSt. Augustine, Trinidad & TobagoWest Indies
| | - Juhi Tiwari
- School of PharmacyJaipur National UniversityJagatpuraJaipurIndia
| | | | | | - Mohit Agrawal
- School of pharmacySuresh Gyan Vihar UniversityJaipurIndia
| | - Anurag Mishra
- School of pharmacySuresh Gyan Vihar UniversityJaipurIndia
| | - Kamal Dua
- Discipline of PharmacyGraduate School of HealthUniversity of Technology SydneySydneyNSWAustralia
- School of Biomedical Sciences and PharmacyUniversity of NewcastleNewcastleNSWAustralia
- School of Pharmaceutical SciencesShoolini UniversitySolanHimachal PradeshIndia
| |
Collapse
|
13
|
Angiotensin IV Receptors Mediate the Cognitive and Cerebrovascular Benefits of Losartan in a Mouse Model of Alzheimer's Disease. J Neurosci 2017; 37:5562-5573. [PMID: 28476949 DOI: 10.1523/jneurosci.0329-17.2017] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 03/30/2017] [Accepted: 04/22/2017] [Indexed: 12/21/2022] Open
Abstract
The use of angiotensin receptor blockers (ARBs) correlates with reduced onset and progression of Alzheimer's disease (AD). The mechanism depicting how ARBs such as losartan restore cerebrovascular and cognitive deficits in AD is unknown. Here, we propose a mechanism underlying losartan's benefits by selectively blocking the effects of angiotensin IV (AngIV) at its receptor (AT4R) with divalinal in mice overexpressing the AD-related Swedish and Indiana mutations of the human amyloid precursor protein (APP mice) and WT mice. Young (3-month-old) mice were treated with losartan (∼10 mg/kg/d, 4 months), followed by intracerebroventricular administration of vehicle or divalinal in the final month of treatment. Spatial learning and memory were assessed using Morris water mazes at 3 and 4 months of losartan treatment. Cerebrovascular reactivity and whisker-evoked neurovascular coupling responses were measured at end point (∼7 months of age), together with biomarkers related to neuronal and vascular oxidative stress (superoxide dismutase-2), neuroinflammation (astroglial and microglial activation), neurogenesis (BrdU-labeled newborn cells), and amyloidosis [soluble amyloid-β (Aβ) species and Aβ plaque load]. Divalinal countered losartan's capacity to rescue spatial learning and memory and blocked losartan's benefits on dilatory function and baseline nitric oxide bioavailability. Divalinal reverted losartan's anti-inflammatory effects, but failed to modify losartan-mediated reductions in oxidative stress. Neither losartan nor divalinal affected arterial blood pressure or significantly altered the amyloid pathology in APP mice. Our findings identify activation of the AngIV/AT4R cascade as the underlying mechanism in losartan's benefits and a target that could restore Aβ-related cognitive and cerebrovascular deficits in AD.SIGNIFICANCE STATEMENT Antihypertensive medications that target the renin angiotensin system, such as angiotensin receptor blockers (ARBs), have been associated with lower incidence and progression of Alzheimer's disease (AD) in cohort studies. However, the manner by which ARBs mediate their beneficial effects is unknown. Here, the angiotensin IV receptor (AT4R) was identified as mediating the cognitive and cerebrovascular rescue of losartan, a commonly prescribed ARB, in a mouse model of AD. The AT4R was further implicated in mediating anti-inflammatory benefits. AT4R-mediated effects were independent from changes in blood pressure, amyloidosis, and oxidative stress. Overall, our results implicate the angiotensin IV/AT4R cascade as a promising candidate for AD intervention.
Collapse
|
14
|
Ongali B, Nicolakakis N, Tong XK, Aboulkassim T, Imboden H, Hamel E. Enalapril Alone or Co-Administered with Losartan Rescues Cerebrovascular Dysfunction, but not Mnemonic Deficits or Amyloidosis in a Mouse Model of Alzheimer's Disease. J Alzheimers Dis 2016; 51:1183-95. [PMID: 26923013 DOI: 10.3233/jad-150868] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The co-administration of angiotensin converting enzyme inhibitors (ACEi) and angiotensin II (AngII) receptor blockers (ARB) that bind angiotensin type 1 receptors (AT1R) may protect from Alzheimer's disease (AD) better than each treatment taken alone. We tested the curative potential of the non brain-penetrant ACEi enalapril (3 mg/kg/day) administered for 3 months either alone or in combination with the brain penetrant ARB losartan (10 mg/kg/day) in aged (∼15 months) transgenic mice overexpressing a mutated form of the human amyloid-β protein precursor (AβPP, thereafter APP mice). We studied cerebrovascular function, protein levels of oxidative stress markers (superoxide dismutases SOD1, SOD2 and the NADPH oxidase subunit p67phox), amyloid-β (Aβ) pathology, astrogliosis, cholinergic innervation, AT1R and angiotensin IV receptor (AT4R) levels, together with cognitive performance. Both treatments normalized cerebrovascular reactivity and p67phox protein levels, but they did not reduce the cerebrovascular levels of SOD1. Combined treatment normalized cerebrovascular SOD2 levels, significantly attenuated astrogliosis, but did not reduce the increased levels of cerebrovascular AT1R. Yet, combined therapy enhanced thioflavin-S labeled Aβ plaque burden, a tendency not significant when Aβ1 - 42 plaque load was considered. None of the treatments rescued cognitive deficits, cortical AT4R or cholinergic innervation. We conclude that both treatments normalized cerebrovascular function by inhibiting the AngII-induced oxidative stress cascade, and that the positive effects of the combined therapy on astrogliosis were likely due to the ability of losartan to enter brain parenchyma. However, enalapril did not potentiate, and may even dampen, the reported cognitive benefits of losartan, raising caution when selecting the most appropriate antihypertensive therapy in AD patients.
Collapse
Affiliation(s)
- Brice Ongali
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Nektaria Nicolakakis
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Xing-Kang Tong
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Tahar Aboulkassim
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Hans Imboden
- Institute of Cell Biology, University of Bern, Switzerland
| | - Edith Hamel
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, QC, Canada
| |
Collapse
|
15
|
Hamel E, Royea J, Ongali B, Tong XK. Neurovascular and Cognitive failure in Alzheimer's Disease: Benefits of Cardiovascular Therapy. Cell Mol Neurobiol 2016; 36:219-32. [PMID: 26993506 DOI: 10.1007/s10571-015-0285-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 10/06/2015] [Indexed: 12/19/2022]
Abstract
Alzheimer's disease (AD) is a multifactorial and multifaceted disease for which we currently have very little to offer since there is no curative therapy, with only limited disease-modifying drugs. Recent studies in AD mouse models that recapitulate the amyloid-β (Aβ) pathology converge to demonstrate that it is possible to salvage cerebrovascular function with a variety of drugs and, particularly, therapies used to treat cardiovascular diseases such as hypercholesterolemia and hypertension. These drugs can reestablish dilatory function mediated by various endothelial and smooth muscle ion channels as well as nitric oxide availability, benefits that result in normalized brain perfusion. These cerebrovascular benefits would favor brain perfusion, which may help maintain neuronal function and, possibly, delay cognitive failure. However, restoring cerebrovascular function in AD mouse models was not necessarily accompanied by rescue of cognitive deficits related to spatial learning and memory. The results with cardiovascular therapies rather suggest that drugs originally designed to treat cardiovascular diseases that concurrently restore cerebrovascular and cognitive function do so through their pleiotropic effects. Specifically, recent findings suggest that these drugs act directly on brain cells and neuronal pathways involved in memory formation, hence, working simultaneously albeit independently on neuronal and vascular targets. These findings may help select medications for patients with cardiovascular diseases at risk of developing AD with increasing age. Further, they may identify molecular targets for recovering memory pathways that bear potential for new therapeutic avenues.
Collapse
Affiliation(s)
- Edith Hamel
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, 3801 University Street, Suite 748, Montréal, QC, H3A 2B4, Canada.
| | - Jessika Royea
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, 3801 University Street, Suite 748, Montréal, QC, H3A 2B4, Canada
| | - Brice Ongali
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, 3801 University Street, Suite 748, Montréal, QC, H3A 2B4, Canada
| | - Xin-Kang Tong
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, 3801 University Street, Suite 748, Montréal, QC, H3A 2B4, Canada
| |
Collapse
|
16
|
Diwakarla S, Nylander E, Grönbladh A, Vanga SR, Khan YS, Gutiérrez-de-Terán H, Ng L, Pham V, Sävmarker J, Lundbäck T, Jenmalm-Jensen A, Andersson H, Engen K, Rosenström U, Larhed M, Åqvist J, Chai SY, Hallberg M. Binding to and Inhibition of Insulin-Regulated Aminopeptidase by Macrocyclic Disulfides Enhances Spine Density. Mol Pharmacol 2016; 89:413-24. [PMID: 26769413 DOI: 10.1124/mol.115.102533] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 01/13/2016] [Indexed: 01/28/2023] Open
Abstract
Angiotensin IV (Ang IV) and related peptide analogs, as well as nonpeptide inhibitors of insulin-regulated aminopeptidase (IRAP), have previously been shown to enhance memory and cognition in animal models. Furthermore, the endogenous IRAP substrates oxytocin and vasopressin are known to facilitate learning and memory. In this study, the two recently synthesized 13-membered macrocyclic competitive IRAP inhibitors HA08 and HA09, which were designed to mimic the N terminus of oxytocin and vasopressin, were assessed and compared based on their ability to bind to the IRAP active site, and alter dendritic spine density in rat hippocampal primary cultures. The binding modes of the IRAP inhibitors HA08, HA09, and of Ang IV in either the extended or γ-turn conformation at the C terminus to human IRAP were predicted by docking and molecular dynamics simulations. The binding free energies calculated with the linear interaction energy method, which are in excellent agreement with experimental data and simulations, have been used to explain the differences in activities of the IRAP inhibitors, both of which are structurally very similar, but differ only with regard to one stereogenic center. In addition, we show that HA08, which is 100-fold more potent than the epimer HA09, can enhance dendritic spine number and alter morphology, a process associated with memory facilitation. Therefore, HA08, one of the most potent IRAP inhibitors known today, may serve as a suitable starting point for medicinal chemistry programs aided by MD simulations aimed at discovering more drug-like cognitive enhancers acting via augmenting synaptic plasticity.
Collapse
Affiliation(s)
- Shanti Diwakarla
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence (S.D., E.N., A.G., M.H.), Department of Cell and Molecular Biology (S.R.V., Y.S.K., H.G.T., J.A.), The Beijer Laboratory, Department of Medicinal Chemistry (J.S.), Department of Medicinal Chemistry (H.A., K.E., U.R.), Science for Life Laboratory, Department of Medicinal Chemistry (M.L.), BMC, Uppsala University, Uppsala, Sweden; Chemical Biology Consortium Sweden, Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medicinal Biochemistry and Biophysics (T.L., A.J.), Karolinska Institute, Sweden; and Biomedicine Discovery Institute, Department of Physiology (L.N., V.P., S.Y.C.), Monash University, Melbourne, Australia
| | - Erik Nylander
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence (S.D., E.N., A.G., M.H.), Department of Cell and Molecular Biology (S.R.V., Y.S.K., H.G.T., J.A.), The Beijer Laboratory, Department of Medicinal Chemistry (J.S.), Department of Medicinal Chemistry (H.A., K.E., U.R.), Science for Life Laboratory, Department of Medicinal Chemistry (M.L.), BMC, Uppsala University, Uppsala, Sweden; Chemical Biology Consortium Sweden, Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medicinal Biochemistry and Biophysics (T.L., A.J.), Karolinska Institute, Sweden; and Biomedicine Discovery Institute, Department of Physiology (L.N., V.P., S.Y.C.), Monash University, Melbourne, Australia
| | - Alfhild Grönbladh
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence (S.D., E.N., A.G., M.H.), Department of Cell and Molecular Biology (S.R.V., Y.S.K., H.G.T., J.A.), The Beijer Laboratory, Department of Medicinal Chemistry (J.S.), Department of Medicinal Chemistry (H.A., K.E., U.R.), Science for Life Laboratory, Department of Medicinal Chemistry (M.L.), BMC, Uppsala University, Uppsala, Sweden; Chemical Biology Consortium Sweden, Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medicinal Biochemistry and Biophysics (T.L., A.J.), Karolinska Institute, Sweden; and Biomedicine Discovery Institute, Department of Physiology (L.N., V.P., S.Y.C.), Monash University, Melbourne, Australia
| | - Sudarsana Reddy Vanga
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence (S.D., E.N., A.G., M.H.), Department of Cell and Molecular Biology (S.R.V., Y.S.K., H.G.T., J.A.), The Beijer Laboratory, Department of Medicinal Chemistry (J.S.), Department of Medicinal Chemistry (H.A., K.E., U.R.), Science for Life Laboratory, Department of Medicinal Chemistry (M.L.), BMC, Uppsala University, Uppsala, Sweden; Chemical Biology Consortium Sweden, Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medicinal Biochemistry and Biophysics (T.L., A.J.), Karolinska Institute, Sweden; and Biomedicine Discovery Institute, Department of Physiology (L.N., V.P., S.Y.C.), Monash University, Melbourne, Australia
| | - Yasmin Shamsudin Khan
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence (S.D., E.N., A.G., M.H.), Department of Cell and Molecular Biology (S.R.V., Y.S.K., H.G.T., J.A.), The Beijer Laboratory, Department of Medicinal Chemistry (J.S.), Department of Medicinal Chemistry (H.A., K.E., U.R.), Science for Life Laboratory, Department of Medicinal Chemistry (M.L.), BMC, Uppsala University, Uppsala, Sweden; Chemical Biology Consortium Sweden, Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medicinal Biochemistry and Biophysics (T.L., A.J.), Karolinska Institute, Sweden; and Biomedicine Discovery Institute, Department of Physiology (L.N., V.P., S.Y.C.), Monash University, Melbourne, Australia
| | - Hugo Gutiérrez-de-Terán
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence (S.D., E.N., A.G., M.H.), Department of Cell and Molecular Biology (S.R.V., Y.S.K., H.G.T., J.A.), The Beijer Laboratory, Department of Medicinal Chemistry (J.S.), Department of Medicinal Chemistry (H.A., K.E., U.R.), Science for Life Laboratory, Department of Medicinal Chemistry (M.L.), BMC, Uppsala University, Uppsala, Sweden; Chemical Biology Consortium Sweden, Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medicinal Biochemistry and Biophysics (T.L., A.J.), Karolinska Institute, Sweden; and Biomedicine Discovery Institute, Department of Physiology (L.N., V.P., S.Y.C.), Monash University, Melbourne, Australia
| | - Leelee Ng
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence (S.D., E.N., A.G., M.H.), Department of Cell and Molecular Biology (S.R.V., Y.S.K., H.G.T., J.A.), The Beijer Laboratory, Department of Medicinal Chemistry (J.S.), Department of Medicinal Chemistry (H.A., K.E., U.R.), Science for Life Laboratory, Department of Medicinal Chemistry (M.L.), BMC, Uppsala University, Uppsala, Sweden; Chemical Biology Consortium Sweden, Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medicinal Biochemistry and Biophysics (T.L., A.J.), Karolinska Institute, Sweden; and Biomedicine Discovery Institute, Department of Physiology (L.N., V.P., S.Y.C.), Monash University, Melbourne, Australia
| | - Vi Pham
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence (S.D., E.N., A.G., M.H.), Department of Cell and Molecular Biology (S.R.V., Y.S.K., H.G.T., J.A.), The Beijer Laboratory, Department of Medicinal Chemistry (J.S.), Department of Medicinal Chemistry (H.A., K.E., U.R.), Science for Life Laboratory, Department of Medicinal Chemistry (M.L.), BMC, Uppsala University, Uppsala, Sweden; Chemical Biology Consortium Sweden, Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medicinal Biochemistry and Biophysics (T.L., A.J.), Karolinska Institute, Sweden; and Biomedicine Discovery Institute, Department of Physiology (L.N., V.P., S.Y.C.), Monash University, Melbourne, Australia
| | - Jonas Sävmarker
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence (S.D., E.N., A.G., M.H.), Department of Cell and Molecular Biology (S.R.V., Y.S.K., H.G.T., J.A.), The Beijer Laboratory, Department of Medicinal Chemistry (J.S.), Department of Medicinal Chemistry (H.A., K.E., U.R.), Science for Life Laboratory, Department of Medicinal Chemistry (M.L.), BMC, Uppsala University, Uppsala, Sweden; Chemical Biology Consortium Sweden, Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medicinal Biochemistry and Biophysics (T.L., A.J.), Karolinska Institute, Sweden; and Biomedicine Discovery Institute, Department of Physiology (L.N., V.P., S.Y.C.), Monash University, Melbourne, Australia
| | - Thomas Lundbäck
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence (S.D., E.N., A.G., M.H.), Department of Cell and Molecular Biology (S.R.V., Y.S.K., H.G.T., J.A.), The Beijer Laboratory, Department of Medicinal Chemistry (J.S.), Department of Medicinal Chemistry (H.A., K.E., U.R.), Science for Life Laboratory, Department of Medicinal Chemistry (M.L.), BMC, Uppsala University, Uppsala, Sweden; Chemical Biology Consortium Sweden, Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medicinal Biochemistry and Biophysics (T.L., A.J.), Karolinska Institute, Sweden; and Biomedicine Discovery Institute, Department of Physiology (L.N., V.P., S.Y.C.), Monash University, Melbourne, Australia
| | - Annika Jenmalm-Jensen
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence (S.D., E.N., A.G., M.H.), Department of Cell and Molecular Biology (S.R.V., Y.S.K., H.G.T., J.A.), The Beijer Laboratory, Department of Medicinal Chemistry (J.S.), Department of Medicinal Chemistry (H.A., K.E., U.R.), Science for Life Laboratory, Department of Medicinal Chemistry (M.L.), BMC, Uppsala University, Uppsala, Sweden; Chemical Biology Consortium Sweden, Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medicinal Biochemistry and Biophysics (T.L., A.J.), Karolinska Institute, Sweden; and Biomedicine Discovery Institute, Department of Physiology (L.N., V.P., S.Y.C.), Monash University, Melbourne, Australia
| | - Hanna Andersson
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence (S.D., E.N., A.G., M.H.), Department of Cell and Molecular Biology (S.R.V., Y.S.K., H.G.T., J.A.), The Beijer Laboratory, Department of Medicinal Chemistry (J.S.), Department of Medicinal Chemistry (H.A., K.E., U.R.), Science for Life Laboratory, Department of Medicinal Chemistry (M.L.), BMC, Uppsala University, Uppsala, Sweden; Chemical Biology Consortium Sweden, Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medicinal Biochemistry and Biophysics (T.L., A.J.), Karolinska Institute, Sweden; and Biomedicine Discovery Institute, Department of Physiology (L.N., V.P., S.Y.C.), Monash University, Melbourne, Australia
| | - Karin Engen
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence (S.D., E.N., A.G., M.H.), Department of Cell and Molecular Biology (S.R.V., Y.S.K., H.G.T., J.A.), The Beijer Laboratory, Department of Medicinal Chemistry (J.S.), Department of Medicinal Chemistry (H.A., K.E., U.R.), Science for Life Laboratory, Department of Medicinal Chemistry (M.L.), BMC, Uppsala University, Uppsala, Sweden; Chemical Biology Consortium Sweden, Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medicinal Biochemistry and Biophysics (T.L., A.J.), Karolinska Institute, Sweden; and Biomedicine Discovery Institute, Department of Physiology (L.N., V.P., S.Y.C.), Monash University, Melbourne, Australia
| | - Ulrika Rosenström
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence (S.D., E.N., A.G., M.H.), Department of Cell and Molecular Biology (S.R.V., Y.S.K., H.G.T., J.A.), The Beijer Laboratory, Department of Medicinal Chemistry (J.S.), Department of Medicinal Chemistry (H.A., K.E., U.R.), Science for Life Laboratory, Department of Medicinal Chemistry (M.L.), BMC, Uppsala University, Uppsala, Sweden; Chemical Biology Consortium Sweden, Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medicinal Biochemistry and Biophysics (T.L., A.J.), Karolinska Institute, Sweden; and Biomedicine Discovery Institute, Department of Physiology (L.N., V.P., S.Y.C.), Monash University, Melbourne, Australia
| | - Mats Larhed
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence (S.D., E.N., A.G., M.H.), Department of Cell and Molecular Biology (S.R.V., Y.S.K., H.G.T., J.A.), The Beijer Laboratory, Department of Medicinal Chemistry (J.S.), Department of Medicinal Chemistry (H.A., K.E., U.R.), Science for Life Laboratory, Department of Medicinal Chemistry (M.L.), BMC, Uppsala University, Uppsala, Sweden; Chemical Biology Consortium Sweden, Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medicinal Biochemistry and Biophysics (T.L., A.J.), Karolinska Institute, Sweden; and Biomedicine Discovery Institute, Department of Physiology (L.N., V.P., S.Y.C.), Monash University, Melbourne, Australia
| | - Johan Åqvist
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence (S.D., E.N., A.G., M.H.), Department of Cell and Molecular Biology (S.R.V., Y.S.K., H.G.T., J.A.), The Beijer Laboratory, Department of Medicinal Chemistry (J.S.), Department of Medicinal Chemistry (H.A., K.E., U.R.), Science for Life Laboratory, Department of Medicinal Chemistry (M.L.), BMC, Uppsala University, Uppsala, Sweden; Chemical Biology Consortium Sweden, Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medicinal Biochemistry and Biophysics (T.L., A.J.), Karolinska Institute, Sweden; and Biomedicine Discovery Institute, Department of Physiology (L.N., V.P., S.Y.C.), Monash University, Melbourne, Australia
| | - Siew Yeen Chai
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence (S.D., E.N., A.G., M.H.), Department of Cell and Molecular Biology (S.R.V., Y.S.K., H.G.T., J.A.), The Beijer Laboratory, Department of Medicinal Chemistry (J.S.), Department of Medicinal Chemistry (H.A., K.E., U.R.), Science for Life Laboratory, Department of Medicinal Chemistry (M.L.), BMC, Uppsala University, Uppsala, Sweden; Chemical Biology Consortium Sweden, Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medicinal Biochemistry and Biophysics (T.L., A.J.), Karolinska Institute, Sweden; and Biomedicine Discovery Institute, Department of Physiology (L.N., V.P., S.Y.C.), Monash University, Melbourne, Australia
| | - Mathias Hallberg
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence (S.D., E.N., A.G., M.H.), Department of Cell and Molecular Biology (S.R.V., Y.S.K., H.G.T., J.A.), The Beijer Laboratory, Department of Medicinal Chemistry (J.S.), Department of Medicinal Chemistry (H.A., K.E., U.R.), Science for Life Laboratory, Department of Medicinal Chemistry (M.L.), BMC, Uppsala University, Uppsala, Sweden; Chemical Biology Consortium Sweden, Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medicinal Biochemistry and Biophysics (T.L., A.J.), Karolinska Institute, Sweden; and Biomedicine Discovery Institute, Department of Physiology (L.N., V.P., S.Y.C.), Monash University, Melbourne, Australia
| |
Collapse
|
17
|
Chow LH, Tao PL, Chen YH, Lin YH, Huang EYK. Angiotensin IV possibly acts through PKMzeta in the hippocampus to regulate cognitive memory in rats. Neuropeptides 2015; 53:1-10. [PMID: 26412453 DOI: 10.1016/j.npep.2015.09.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 08/10/2015] [Accepted: 09/10/2015] [Indexed: 10/23/2022]
Abstract
Ang IV is an endogenous peptide generated from the degradation of angiotensin II. Ang IV was found to enhance learning and memory in CNS. PKMzeta was identified to be a fragment of PKCzeta (protein kinase Czeta). Its continuous activation was demonstrated to be correlated with the formation of memory in the hippocampus. Therefore, we investigated whether PKMzeta participates in the effects of Ang IV on memory. We first examined the effect of Ang IV on non-spatial memory/cognition in modified object recognition test in rats. Our data showed that Ang IV could increase the exploration time on novel object. The co-administration of ZIP (PKMzeta inhibitor) with Ang IV significantly blocked the effect by Ang IV. The effects of Ang IV on hippocampal LTP at the CA1 region were also evaluated. Ang IV significantly increased the amplitude and slope of the EPSPs, which was consistent with other reports. Surprisingly, instead of potentiating LTP, Ang IV caused a failed maintenance of LTP. Moreover, there was no quantitative change in PKMzeta induced by Ang IV and/or ZIP after behavioral experiments. Taken together, our data re-confirmed the finding of the positive effect of Ang IV to enhance memory/cognition. The increased strength of EPSPs with Ang IV could also have certain functional relevance. Since the behavioral results suggested the involvement of PKMzeta, we hypothesized that the enhancement of memory/cognition by Ang IV may rely on an increase in PKMzeta activity. Overall, the present study provided important advances in our understanding of the action of Ang IV in the hippocampus.
Collapse
Affiliation(s)
- Lok-Hi Chow
- Department of Pharmacology, National Defense Medical Center, Nei-Hu, 114 Taipei, Taiwan, ROC; Department of Anesthesiology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC; Department of Anesthesiology, School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
| | - Pao-Luh Tao
- Center for Neuropsychiatric Research, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County 350, Taiwan, ROC
| | - Yuan-Hao Chen
- Department of Neurological Surgery, Tri-Service General Hospital, National Defense Medical Center, Nei-Hu, 114 Taipei, Taiwan, ROC
| | - Yu-Hui Lin
- Department of Pharmacology, National Defense Medical Center, Nei-Hu, 114 Taipei, Taiwan, ROC
| | - Eagle Yi-Kung Huang
- Department of Pharmacology, National Defense Medical Center, Nei-Hu, 114 Taipei, Taiwan, ROC.
| |
Collapse
|
18
|
Karnik SS, Unal H, Kemp JR, Tirupula KC, Eguchi S, Vanderheyden PML, Thomas WG. International Union of Basic and Clinical Pharmacology. XCIX. Angiotensin Receptors: Interpreters of Pathophysiological Angiotensinergic Stimuli [corrected]. Pharmacol Rev 2015; 67:754-819. [PMID: 26315714 PMCID: PMC4630565 DOI: 10.1124/pr.114.010454] [Citation(s) in RCA: 215] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The renin angiotensin system (RAS) produced hormone peptides regulate many vital body functions. Dysfunctional signaling by receptors for RAS peptides leads to pathologic states. Nearly half of humanity today would likely benefit from modern drugs targeting these receptors. The receptors for RAS peptides consist of three G-protein-coupled receptors—the angiotensin II type 1 receptor (AT1 receptor), the angiotensin II type 2 receptor (AT2 receptor), the MAS receptor—and a type II trans-membrane zinc protein—the candidate angiotensin IV receptor (AngIV binding site). The prorenin receptor is a relatively new contender for consideration, but is not included here because the role of prorenin receptor as an independent endocrine mediator is presently unclear. The full spectrum of biologic characteristics of these receptors is still evolving, but there is evidence establishing unique roles of each receptor in cardiovascular, hemodynamic, neurologic, renal, and endothelial functions, as well as in cell proliferation, survival, matrix-cell interaction, and inflammation. Therapeutic agents targeted to these receptors are either in active use in clinical intervention of major common diseases or under evaluation for repurposing in many other disorders. Broad-spectrum influence these receptors produce in complex pathophysiological context in our body highlights their role as precise interpreters of distinctive angiotensinergic peptide cues. This review article summarizes findings published in the last 15 years on the structure, pharmacology, signaling, physiology, and disease states related to angiotensin receptors. We also discuss the challenges the pharmacologist presently faces in formally accepting newer members as established angiotensin receptors and emphasize necessary future developments.
Collapse
Affiliation(s)
- Sadashiva S Karnik
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Hamiyet Unal
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Jacqueline R Kemp
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Kalyan C Tirupula
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Satoru Eguchi
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Patrick M L Vanderheyden
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| | - Walter G Thomas
- Department of Molecular Cardiology, Lerner Research Institute of Cleveland Clinic, Cleveland, Ohio (S.S.K., H.U., J.R.K., K.C.T.); Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania (S.E.); Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium (P.M.L.V.); and Department of General Physiology, School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia (W.G.T.)
| |
Collapse
|
19
|
Wright JW, Kawas LH, Harding JW. The development of small molecule angiotensin IV analogs to treat Alzheimer's and Parkinson's diseases. Prog Neurobiol 2014; 125:26-46. [PMID: 25455861 DOI: 10.1016/j.pneurobio.2014.11.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 11/17/2014] [Accepted: 11/19/2014] [Indexed: 02/07/2023]
Abstract
Alzheimer's (AD) and Parkinson's (PD) diseases are neurodegenerative diseases presently without effective drug treatments. AD is characterized by general cognitive impairment, difficulties with memory consolidation and retrieval, and with advanced stages episodes of agitation and anger. AD is increasing in frequency as life expectancy increases. Present FDA approved medications do little to slow disease progression and none address the underlying progressive loss of synaptic connections and neurons. New drug design approaches are needed beyond cholinesterase inhibitors and N-methyl-d-aspartate receptor antagonists. Patients with PD experience the symptomatic triad of bradykinesis, tremor-at-rest, and rigidity with the possibility of additional non-motor symptoms including sleep disturbances, depression, dementia, and autonomic nervous system failure. This review summarizes available information regarding the role of the brain renin-angiotensin system (RAS) in learning and memory and motor functions, with particular emphasis on research results suggesting a link between angiotensin IV (AngIV) interacting with the AT4 receptor subtype. Currently there is controversy over the identity of this AT4 receptor protein. Albiston and colleagues have offered convincing evidence that it is the insulin-regulated aminopeptidase (IRAP). Recently members of our laboratory have presented evidence that the brain AngIV/AT4 receptor system coincides with the brain hepatocyte growth factor/c-Met receptor system. In an effort to resolve this issue we have synthesized a number of small molecule AngIV-based compounds that are metabolically stable, penetrate the blood-brain barrier, and facilitate compromised memory and motor systems. These research efforts are described along with details concerning a recently synthesized molecule, Dihexa that shows promise in overcoming memory and motor dysfunctions by augmenting synaptic connectivity via the formation of new functional synapses.
Collapse
Affiliation(s)
- John W Wright
- Departments of Psychology, Integrative Physiology and Neuroscience and Program in Biotechnology, Washington State University, Pullman, WA 99164-4820, USA; M3 Biotechnology, Inc., 4000 Mason Rd Suite 300, Box 352141, Seattle, WA 98195-2141, USA.
| | - Leen H Kawas
- Departments of Psychology, Integrative Physiology and Neuroscience and Program in Biotechnology, Washington State University, Pullman, WA 99164-4820, USA; M3 Biotechnology, Inc., 4000 Mason Rd Suite 300, Box 352141, Seattle, WA 98195-2141, USA
| | - Joseph W Harding
- Departments of Psychology, Integrative Physiology and Neuroscience and Program in Biotechnology, Washington State University, Pullman, WA 99164-4820, USA; M3 Biotechnology, Inc., 4000 Mason Rd Suite 300, Box 352141, Seattle, WA 98195-2141, USA
| |
Collapse
|
20
|
Benoist CC, Kawas LH, Zhu M, Tyson KA, Stillmaker L, Appleyard SM, Wright JW, Wayman GA, Harding JW. The procognitive and synaptogenic effects of angiotensin IV-derived peptides are dependent on activation of the hepatocyte growth factor/c-met system. J Pharmacol Exp Ther 2014; 351:390-402. [PMID: 25187433 DOI: 10.1124/jpet.114.218735] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
A subset of angiotensin IV (AngIV)-related molecules are known to possess procognitive/antidementia properties and have been considered as templates for potential therapeutics. However, this potential has not been realized because of two factors: 1) a lack of blood-brain barrier-penetrant analogs, and 2) the absence of a validated mechanism of action. The pharmacokinetic barrier has recently been overcome with the synthesis of the orally active, blood-brain barrier-permeable analog N-hexanoic-tyrosine-isoleucine-(6) aminohexanoic amide (dihexa). Therefore, the goal of this study was to elucidate the mechanism that underlies dihexa's procognitive activity. Here, we demonstrate that dihexa binds with high affinity to hepatocyte growth factor (HGF) and both dihexa and its parent compound Norleucine 1-AngIV (Nle(1)-AngIV) induce c-Met phosphorylation in the presence of subthreshold concentrations of HGF and augment HGF-dependent cell scattering. Further, dihexa and Nle(1)-AngIV induce hippocampal spinogenesis and synaptogenesis similar to HGF itself. These actions were inhibited by an HGF antagonist and a short hairpin RNA directed at c-Met. Most importantly, the procognitive/antidementia capacity of orally delivered dihexa was blocked by an HGF antagonist delivered intracerebroventricularly as measured using the Morris water maze task of spatial learning.
Collapse
Affiliation(s)
- Caroline C Benoist
- Department of Integrative Physiology and Neuroscience (C.C.B., L.H.K., M.Z., K.A.T., L.S., S.M.A., J.W.W., G.A.W., J.W.H.) and Department of Psychology (J.W.W., J.W.H.), Washington State University, Pullman, Washington; and M Biotechnology, Inc., Seattle, Washington (L.H.K., J.W.W., J.W.H.)
| | - Leen H Kawas
- Department of Integrative Physiology and Neuroscience (C.C.B., L.H.K., M.Z., K.A.T., L.S., S.M.A., J.W.W., G.A.W., J.W.H.) and Department of Psychology (J.W.W., J.W.H.), Washington State University, Pullman, Washington; and M Biotechnology, Inc., Seattle, Washington (L.H.K., J.W.W., J.W.H.)
| | - Mingyan Zhu
- Department of Integrative Physiology and Neuroscience (C.C.B., L.H.K., M.Z., K.A.T., L.S., S.M.A., J.W.W., G.A.W., J.W.H.) and Department of Psychology (J.W.W., J.W.H.), Washington State University, Pullman, Washington; and M Biotechnology, Inc., Seattle, Washington (L.H.K., J.W.W., J.W.H.)
| | - Katherine A Tyson
- Department of Integrative Physiology and Neuroscience (C.C.B., L.H.K., M.Z., K.A.T., L.S., S.M.A., J.W.W., G.A.W., J.W.H.) and Department of Psychology (J.W.W., J.W.H.), Washington State University, Pullman, Washington; and M Biotechnology, Inc., Seattle, Washington (L.H.K., J.W.W., J.W.H.)
| | - Lori Stillmaker
- Department of Integrative Physiology and Neuroscience (C.C.B., L.H.K., M.Z., K.A.T., L.S., S.M.A., J.W.W., G.A.W., J.W.H.) and Department of Psychology (J.W.W., J.W.H.), Washington State University, Pullman, Washington; and M Biotechnology, Inc., Seattle, Washington (L.H.K., J.W.W., J.W.H.)
| | - Suzanne M Appleyard
- Department of Integrative Physiology and Neuroscience (C.C.B., L.H.K., M.Z., K.A.T., L.S., S.M.A., J.W.W., G.A.W., J.W.H.) and Department of Psychology (J.W.W., J.W.H.), Washington State University, Pullman, Washington; and M Biotechnology, Inc., Seattle, Washington (L.H.K., J.W.W., J.W.H.)
| | - John W Wright
- Department of Integrative Physiology and Neuroscience (C.C.B., L.H.K., M.Z., K.A.T., L.S., S.M.A., J.W.W., G.A.W., J.W.H.) and Department of Psychology (J.W.W., J.W.H.), Washington State University, Pullman, Washington; and M Biotechnology, Inc., Seattle, Washington (L.H.K., J.W.W., J.W.H.)
| | - Gary A Wayman
- Department of Integrative Physiology and Neuroscience (C.C.B., L.H.K., M.Z., K.A.T., L.S., S.M.A., J.W.W., G.A.W., J.W.H.) and Department of Psychology (J.W.W., J.W.H.), Washington State University, Pullman, Washington; and M Biotechnology, Inc., Seattle, Washington (L.H.K., J.W.W., J.W.H.)
| | - Joseph W Harding
- Department of Integrative Physiology and Neuroscience (C.C.B., L.H.K., M.Z., K.A.T., L.S., S.M.A., J.W.W., G.A.W., J.W.H.) and Department of Psychology (J.W.W., J.W.H.), Washington State University, Pullman, Washington; and M Biotechnology, Inc., Seattle, Washington (L.H.K., J.W.W., J.W.H.)
| |
Collapse
|
21
|
Ongali B, Nicolakakis N, Tong XK, Aboulkassim T, Papadopoulos P, Rosa-Neto P, Lecrux C, Imboden H, Hamel E. Angiotensin II type 1 receptor blocker losartan prevents and rescues cerebrovascular, neuropathological and cognitive deficits in an Alzheimer's disease model. Neurobiol Dis 2014; 68:126-36. [PMID: 24807206 DOI: 10.1016/j.nbd.2014.04.018] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 04/17/2014] [Accepted: 04/27/2014] [Indexed: 11/18/2022] Open
Abstract
Angiotensin II (AngII) receptor blockers that bind selectively AngII type 1 (AT1) receptors may protect from Alzheimer's disease (AD). We studied the ability of the AT1 receptor antagonist losartan to cure or prevent AD hallmarks in aged (~18months at endpoint, 3months treatment) or adult (~12months at endpoint, 10months treatment) human amyloid precursor protein (APP) transgenic mice. We tested learning and memory with the Morris water maze, and evaluated neurometabolic and neurovascular coupling using [(18)F]fluoro-2-deoxy-D-glucose-PET and laser Doppler flowmetry responses to whisker stimulation. Cerebrovascular reactivity was assessed with on-line videomicroscopy. We measured protein levels of oxidative stress enzymes (superoxide dismutases SOD1, SOD2 and NADPH oxidase subunit p67phox), and quantified soluble and deposited amyloid-β (Aβ) peptide, glial fibrillary acidic protein (GFAP), AngII receptors AT1 and AT2, angiotensin IV receptor AT4, and cortical cholinergic innervation. In aged APP mice, losartan did not improve learning but it consolidated memory acquisition and recall, and rescued neurovascular and neurometabolic coupling and cerebrovascular dilatory capacity. Losartan normalized cerebrovascular p67phox and SOD2 protein levels and up-regulated those of SOD1. Losartan attenuated astrogliosis, normalized AT1 and AT4 receptor levels, but failed to rescue the cholinergic deficit and the Aβ pathology. Given preventively, losartan protected cognitive function, cerebrovascular reactivity, and AT4 receptor levels. Like in aged APP mice, these benefits occurred without a decrease in soluble Aβ species or plaque load. We conclude that losartan exerts potent preventive and restorative effects on AD hallmarks, possibly by mitigating AT1-initiated oxidative stress and normalizing memory-related AT4 receptors.
Collapse
Affiliation(s)
- Brice Ongali
- Laboratory of Cerebrovascular Research, McGill University, Montréal, QC H3A 2B4, Canada
| | - Nektaria Nicolakakis
- Laboratory of Cerebrovascular Research, McGill University, Montréal, QC H3A 2B4, Canada
| | - Xin-Kang Tong
- Laboratory of Cerebrovascular Research, McGill University, Montréal, QC H3A 2B4, Canada
| | - Tahar Aboulkassim
- Laboratory of Cerebrovascular Research, McGill University, Montréal, QC H3A 2B4, Canada
| | | | - Pedro Rosa-Neto
- Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada; Douglas Hospital Research Centre, McGill University, Montréal, QC H3A 2B4, Canada
| | - Clotilde Lecrux
- Laboratory of Cerebrovascular Research, McGill University, Montréal, QC H3A 2B4, Canada
| | - Hans Imboden
- Institute of Cell Biology, University of Bern, Switzerland
| | - Edith Hamel
- Laboratory of Cerebrovascular Research, McGill University, Montréal, QC H3A 2B4, Canada.
| |
Collapse
|
22
|
Genaro K, Juliano MA, Prado WA, Brandão ML, Martins AR. Effects of angiotensin (5-8) microinfusions into the ventrolateral periaqueductal gray on defensive behaviors in rats. Behav Brain Res 2013; 256:537-44. [PMID: 24041538 DOI: 10.1016/j.bbr.2013.09.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 09/06/2013] [Accepted: 09/10/2013] [Indexed: 10/26/2022]
Abstract
Peptides of the renin-angiotensin system modulate blood pressure and hydro-electrolyte composition. Angiotensin (Ang) receptors are localized in brain areas related to the regulation of autonomic and endocrine control and involved in sensory perception, memory process and behavioral responses. Among these areas, the ventrolateral periaqueductal gray (vlPAG) is one of the most important structures of the neuronal circuitry controlling the autonomic and behavioral components of emotional states. Although Ang II metabolism in the vlPAG forms several Ang-peptides including Ang (5-8), the role of this tetrapeptide in the organization of defensive responses has not yet been described. To address this issue, the purpose of the present study was to determine the effects of intra-vlPAG injections of Ang (5-8) (0.2, 0.4 and 0.8 nmol/0.25 μL) in rats submitted to the elevated plus-maze (EPM) test. Additionally, it was evaluated the effects of intra-vlPAG Ang (5-8) on the expression of conditioned fear, assessed by the fear-potentiated startle and contextual conditioned freezing tests. The results showed that Ang (5-8) produced an intense, dose-related reduction in the entries into and time spent in the open arms of the EPM, decreased direct exploration and increased risk assessment behaviors. Moreover, intra-vlPAG injections of Ang (5-8) before the test session promoted pro-aversive effects in the FPS and enhanced contextual freezing. Taken together, these results point out to an important anxiogenic-like action for Ang (5-8) in the mediation of defensive behaviors organized in the vlPAG.
Collapse
Affiliation(s)
- Karina Genaro
- Universidade Federal do Triângulo Mineiro, Instituto de Ciências Biológicas, Uberaba, MG, Brazil; Universidade de São Paulo, Departamento de Farmacologia, Ribeirão Preto, SP, Brazil; Instituto de Neurociências e Comportamento, INeC, Ribeirão Preto, SP, Brazil.
| | | | | | | | | |
Collapse
|
23
|
Tota S, Goel R, Pachauri SD, Rajasekar N, Najmi AK, Hanif K, Nath C. Effect of angiotensin II on spatial memory, cerebral blood flow, cholinergic neurotransmission, and brain derived neurotrophic factor in rats. Psychopharmacology (Berl) 2013. [PMID: 23192311 DOI: 10.1007/s00213-012-2913-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
RATIONAL Studies have shown the involvement of angiotensin II (Ang II) in neurobehavioral aspects, but the exact role of Ang II in memory is still ambiguous. OBJECTIVE This study explored the effect of central Ang II on spatial memory along with cholinergic neurotransmission, brain energy metabolism, cerebral blood flow (CBF), and brain-derived neurotrophic factor (BDNF) in rats. METHODS Spatial memory was evaluated by Morris water maze (MWM) after Ang II (ICV) administration in male Sprague-Dawley rats. CBF was measured by laser Doppler flowmetry. Oxidative stress adenosine triphosphate (ATP), BDNF, acetylcholinesterase (AChE), and acetylcholine (ACh) were estimated in the cortex and hippocampus at 1, 24, and 48 h after Ang II administration. The effect of AT1 and AT2 receptor blocker (candesartan and PD123,319, respectively), AChE inhibitor (donepezil), and antioxidant melatonin was studied on memory, CBF, and biochemical parameters. RESULTS Ang II caused spatial memory impairment by affecting acquisition, consolidation, and recall in the MWM test along with a significant reduction in CBF. Ang II significantly reduced ACh level and caused oxidative stress in the rat brain 1 h post-injection. No significant change was observed in BDNF, AChE, and ATP level. Candesartan and donepezil prevented Ang II-induced memory impairment, reduction in CBF and ACh level. However, PD123,319 and melatonin failed to prevent Ang II-induced memory impairment but improved CBF partially. CONCLUSION This study suggests that Ang II, via the AT1 receptor, affects spatial memory formation, CBF, and ACh level while AT2 receptor has no significant role.
Collapse
Affiliation(s)
- Santoshkumar Tota
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | | | | | | | | | | | | |
Collapse
|
24
|
Paris JJ, Eans SO, Mizrachi E, Reilley KJ, Ganno ML, McLaughlin JP. Central administration of angiotensin IV rapidly enhances novel object recognition among mice. Neuropharmacology 2013; 70:247-53. [PMID: 23416700 DOI: 10.1016/j.neuropharm.2013.01.025] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Revised: 01/24/2013] [Accepted: 01/25/2013] [Indexed: 12/31/2022]
Abstract
Angiotensin IV (Val(1)-Tyr(2)-Ile(3)-His(4)-Pro(5)-Phe(6)) has demonstrated potential cognitive-enhancing effects. The present investigation assessed and characterized: (1) dose-dependency of angiotensin IV's cognitive enhancement in a C57BL/6J mouse model of novel object recognition, (2) the time-course for these effects, (3) the identity of residues in the hexapeptide important to these effects and (4) the necessity of actions at angiotensin IV receptors for procognitive activity. Assessment of C57BL/6J mice in a novel object recognition task demonstrated that prior administration of angiotensin IV (0.1, 1.0, or 10.0, but not 0.01 nmol, i.c.v.) significantly enhanced novel object recognition in a dose-dependent manner. These effects were time dependent, with improved novel object recognition observed when angiotensin IV (0.1 nmol, i.c.v.) was administered 10 or 20, but not 30 min prior to the onset of the novel object recognition testing. An alanine scan of the angiotensin IV peptide revealed that replacement of the Val(1), Ile(3), His(4), or Phe(6) residues with Ala attenuated peptide-induced improvements in novel object recognition, whereas Tyr(2) or Pro(5) replacement did not significantly affect performance. Administration of the angiotensin IV receptor antagonist, divalinal-Ang IV (20 nmol, i.c.v.), reduced (but did not abolish) novel object recognition; however, this antagonist completely blocked the procognitive effects of angiotensin IV (0.1 nmol, i.c.v.) in this task. Rotorod testing demonstrated no locomotor effects with any angiotensin IV or divalinal-Ang IV dose tested. These data demonstrate that angiotensin IV produces a rapid enhancement of associative learning and memory performance in a mouse model that was dependent on the angiotensin IV receptor.
Collapse
Affiliation(s)
- Jason J Paris
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Parkway, Port Saint Lucie, FL 34987, USA.
| | | | | | | | | | | |
Collapse
|
25
|
U-shaped relation between plasma oxytocin levels and behavior in the trust game. PLoS One 2012; 7:e51095. [PMID: 23227239 PMCID: PMC3515439 DOI: 10.1371/journal.pone.0051095] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Accepted: 10/29/2012] [Indexed: 11/19/2022] Open
Abstract
Trust underpins much of social and economic exchanges across human societies. In experimental economics, the Trust Game has served as the workhorse for the study of trust in a controlled incentivized setting. Recent evidence using intranasal drug administration, aka ‘sniffing’, suggests that oxytocin (OT) can function as a social hormone facilitating trust and other affiliative behaviors. Here we hypothesized that baseline plasma OT is a biomarker that partially predicts the degree of trust and trustworthiness observed in the trust game. Using a large sample of 1,158 participants, we observed a significant U-shaped relationship between plasma OT with the level of trust, and marginally with the level of trustworthiness, especially among males. Specifically, subjects with more extreme levels of plasma OT were more likely to be trusting as well as trustworthy than those with moderate levels of plasma OT. Our results contribute to a deeper understanding of the biological basis of human trust and underscore the usefulness of peripheral plasma OT measures in characterizing human social behavior.
Collapse
|
26
|
Wright JW, Harding JW. Importance of the brain Angiotensin system in Parkinson's disease. PARKINSON'S DISEASE 2012; 2012:860923. [PMID: 23213621 PMCID: PMC3503402 DOI: 10.1155/2012/860923] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Revised: 10/01/2012] [Accepted: 10/02/2012] [Indexed: 11/17/2022]
Abstract
Parkinson's disease (PD) has become a major health problem affecting 1.5% of the world's population over 65 years of age. As life expectancy has increased so has the occurrence of PD. The primary direct consequence of this disease is the loss of dopaminergic (DA) neurons in the substantia nigra and striatum. As the intensity of motor dysfunction increases, the symptomatic triad of bradykinesia, tremors-at-rest, and rigidity occur. Progressive neurodegeneration may also impact non-DA neurotransmitter systems including cholinergic, noradrenergic, and serotonergic, often leading to the development of depression, sleep disturbances, dementia, and autonomic nervous system failure. L-DOPA is the most efficacious oral delivery treatment for controlling motor symptoms; however, this approach is ineffective regarding nonmotor symptoms. New treatment strategies are needed designed to provide neuroprotection and encourage neurogenesis and synaptogenesis to slow or reverse this disease process. The hepatocyte growth factor (HGF)/c-Met receptor system is a member of the growth factor family and has been shown to protect against degeneration of DA neurons in animal models. Recently, small angiotensin-based blood-brain barrier penetrant mimetics have been developed that activate this HGF/c-Met system. These compounds may offer a new and novel approach to the treatment of Parkinson's disease.
Collapse
Affiliation(s)
- John W. Wright
- Departments of Psychology, Veterinary and Comparative Anatomy, Pharmacology, and Physiology and Programs in Neuroscience and Biotechnology, Washington State University, P.O. Box 644820, Pullman, WA 99164-4820, USA
| | - Joseph W. Harding
- Departments of Psychology, Veterinary and Comparative Anatomy, Pharmacology, and Physiology and Programs in Neuroscience and Biotechnology, Washington State University, P.O. Box 644820, Pullman, WA 99164-4820, USA
| |
Collapse
|
27
|
Wright JW, Harding JW. The brain renin–angiotensin system: a diversity of functions and implications for CNS diseases. Pflugers Arch 2012; 465:133-51. [DOI: 10.1007/s00424-012-1102-2] [Citation(s) in RCA: 154] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Revised: 03/20/2012] [Accepted: 03/30/2012] [Indexed: 12/14/2022]
|
28
|
Albiston AL, Diwakarla S, Fernando RN, Mountford SJ, Yeatman HR, Morgan B, Pham V, Holien JK, Parker MW, Thompson PE, Chai SY. Identification and development of specific inhibitors for insulin-regulated aminopeptidase as a new class of cognitive enhancers. Br J Pharmacol 2012; 164:37-47. [PMID: 21470200 DOI: 10.1111/j.1476-5381.2011.01402.x] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Two structurally distinct peptides, angiotensin IV and LVV-haemorphin 7, both competitive high-affinity inhibitors of insulin-regulated aminopeptidase (IRAP), were found to enhance aversion-associated and spatial memory in normal rats and to improve performance in a number of memory tasks in rat deficits models. These findings provide compelling support for the development of specific, high-affinity inhibitors of the enzyme as new cognitive enhancing agents. Different classes of IRAP inhibitors have been developed including peptidomimetics and small molecular weight compounds identified through in silico screening with a homology model of the catalytic domain of IRAP. The proof of principal that inhibition of IRAP activity results in facilitation of memory has been obtained by the demonstration that the small-molecule IRAP inhibitors also exhibit memory-enhancing properties.
Collapse
Affiliation(s)
- Anthony L Albiston
- Howard Florey Institute, University of Melbourne, Parkville, Victoria, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Wright JW, Harding JW. Brain renin-angiotensin—A new look at an old system. Prog Neurobiol 2011; 95:49-67. [DOI: 10.1016/j.pneurobio.2011.07.001] [Citation(s) in RCA: 147] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2011] [Revised: 06/27/2011] [Accepted: 07/03/2011] [Indexed: 12/15/2022]
|
30
|
Andersson H, Demaegdt H, Johnsson A, Vauquelin G, Lindeberg G, Hallberg M, Erdélyi M, Karlén A, Hallberg A. Potent Macrocyclic Inhibitors of Insulin-Regulated Aminopeptidase (IRAP) by Olefin Ring-Closing Metathesis. J Med Chem 2011; 54:3779-92. [DOI: 10.1021/jm200036n] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Hanna Andersson
- Department of Medicinal Chemistry, Uppsala University, Box 574, SE-751 23 Uppsala, Sweden
| | - Heidi Demaegdt
- Department of Molecular and Biochemical Pharmacology, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium
| | - Anders Johnsson
- Department of Medicinal Chemistry, Uppsala University, Box 574, SE-751 23 Uppsala, Sweden
| | - Georges Vauquelin
- Department of Molecular and Biochemical Pharmacology, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium
| | - Gunnar Lindeberg
- Department of Medicinal Chemistry, Uppsala University, Box 574, SE-751 23 Uppsala, Sweden
| | - Mathias Hallberg
- Department of Pharmaceutical Biosciences, Uppsala University, Box 591, SE-751 24 Uppsala, Sweden
| | - Máté Erdélyi
- Department of Chemistry, University of Gothenburg, SE-412 96 Gothenburg, Sweden
- Swedish NMR Centre, University of Gothenburg, Box 465, SE-405 30 Gothenburg, Sweden
| | - Anders Karlén
- Department of Medicinal Chemistry, Uppsala University, Box 574, SE-751 23 Uppsala, Sweden
| | - Anders Hallberg
- Department of Medicinal Chemistry, Uppsala University, Box 574, SE-751 23 Uppsala, Sweden
| |
Collapse
|
31
|
Olson ML, Cero IJ. Intrahippocampal Norleucine¹-Angiotensin IV mitigates scopolamine-induced spatial working memory deficits. Peptides 2010; 31:2209-15. [PMID: 20816712 DOI: 10.1016/j.peptides.2010.08.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2010] [Revised: 08/23/2010] [Accepted: 08/24/2010] [Indexed: 11/19/2022]
Abstract
Depletion of cholinergic neurons in the hippocampus has been implicated in memory impairment and Alzheimer's Disease (AD). The brain angiotensin AT₄ receptor is co-localized with cholinergic neurons, and the AT₄ receptor has also been implicated in cognitive processing. The current investigation used the spatial win-shift version of the radial arm maze to determine the involvement of AT₄ receptors in spatial working memory formation. We initially established that intrahippocampal scopolamine significantly impaired the spatial working memory performance of Sprague-Dawley rats in the radial arm maze. We also demonstrated that subsequent intrahippocampal infusions of Norleucine¹-Angiotensin IV (Nle¹-AngIV) significantly prevented the scopolamine-induced deficit. Consistent with previously published data on long-term spatial memory, our findings suggest that activation of AT₄ receptors can compensate for impaired spatial working memory resulting from compromised muscarinic acetylcholine receptor function. We further demonstrate that the hippocampus is a site of action for Nle¹-AngIV-mediated cognitive improvement.
Collapse
Affiliation(s)
- Mikel L Olson
- Department of Psychology, Concordia College, 901 8th St. S., Moorhead, MN 56562, USA.
| | | |
Collapse
|
32
|
Albrecht D. Physiological and pathophysiological functions of different angiotensins in the brain. Br J Pharmacol 2010. [DOI: 10.1111/j.1476-5381.2010.00648.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
|
33
|
Wright JW, Harding JW. The brain angiotensin IV/AT4receptor system as a new target for the treatment of Alzheimer's disease. Drug Dev Res 2009. [DOI: 10.1002/ddr.20328] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
34
|
Braszko JJ. Participation of D 1-4 dopamine receptors in the pro-cognitive effects of angiotensin IV and des-Phe 6 angiotensin IV. Neurosci Biobehav Rev 2009; 34:343-50. [PMID: 19686774 DOI: 10.1016/j.neubiorev.2009.08.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2009] [Revised: 08/10/2009] [Accepted: 08/10/2009] [Indexed: 10/20/2022]
Abstract
Angiotensin IV (Ang IV) and des-Phe(6)Ang IV are naturally occurring neuroactive peptides of the renin-angiotensin system (RAS) involved in memory processing. However, the relevant mechanisms are poorly understood. In this review it is proposed that the pro-cognitive effects of these peptides are, at least partly, mediated by dopamine (DA). Recent studies demonstrated that the improvement of several memory aspects; recall of appetitively and aversively motivated behaviors and learning of spatial tasks by Ang IV and des-Phe(6)Ang IV was abolished, or significantly diminished by behaviorally inactive per se doses of the D(1) and D(2) receptor blockers SCH 23390 (R-[+]-7-chloro-8-hydroxy-3 methyl-1-phenyl-2,3,4,5-tetrahydro-1H-3-benzazepine) and remoxipride, respectively. The D(3) receptor inhibition with nafadotride was almost ineffective but again, the D(4) receptor blockade by L745,870 hydrochloride (3-{[4-(4-chlorophenyl)piperazin-1-yl]methyl}-1H-pyrrolo[2,3-b]pyridine hydrochloride) diminished all, except for spatial memory, improving actions of the peptides. These results suggest that Ang IV and des-Phe(6)Ang IV enhance memory in a brain region-specific manner, dependent on local DA receptor subpopulations and the memory aspects controlled by them. The data reviewed here, demonstrating DA-Ang IV and des-Phe(6)Ang IV interactions in brain, strongly suggest probability of clinically relevant effects of concomitant use of antipsychotic and RAS affecting drugs.
Collapse
Affiliation(s)
- Jan J Braszko
- Department of Clinical Pharmacology, Medical University of Bialystok, Waszyngtona 15 A, 15274 Bialystok, Poland.
| |
Collapse
|
35
|
Wright JW, Harding JW. The angiotensin AT4 receptor subtype as a target for the treatment of memory dysfunction associated with Alzheimer's disease. J Renin Angiotensin Aldosterone Syst 2009; 9:226-37. [PMID: 19126664 DOI: 10.1177/1470320308099084] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Over recent years antihypertensive drugs, particularly angiotensin-converting enzyme (ACE) inhibitors and angiotensin receptor blockers (ARBs), have been reported to have beneficial effects upon cognitive impairment. Such findings suggest that pharmacological manipulation of angiotensin ligands may be of clinical importance in slowing or halting the cognitive deterioration seen in vascular dementia and Alzheimer's disease. The mechanism(s) underlying these improvements in cognitive function remains unclear; however, important leads are emerging. The angiotensin AT4 receptor subtype, discovered by our laboratory in 1992, influences several important behaviours and physiologies, including learning and memory, and may play a role in this cognitive improvement. This review initially describes the therapeutic drugs approved by the Federal Drug Administration and new approaches presently being developed to treat Alzheimer's disease-induced cognitive impairment. Next, the biologically-active angiotensin ligands and their respective receptor subtypes are discussed, followed by the roles of angiotensin II, angiotensin IV, ACE inhibitors and ARBs in cognitive function. We conclude with a working hypothesis concerning the importance of the AT4 receptor subtype as a new potential drug target for the treatment of Alzheimer's disease-associated memory loss.
Collapse
Affiliation(s)
- John W Wright
- Department of Psychology, Veterinary and Comparative Anatomy, Pharmacology, and Physiology, Washington State University, P.O. Box 644820, Pullman, WA 99164-4820, USA.
| | | |
Collapse
|
36
|
Vanderheyden PML. From angiotensin IV binding site to AT4 receptor. Mol Cell Endocrinol 2009; 302:159-66. [PMID: 19071192 DOI: 10.1016/j.mce.2008.11.015] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2008] [Revised: 11/03/2008] [Accepted: 11/03/2008] [Indexed: 12/23/2022]
Abstract
One of the fragments of the cardiovascular hormone Angiotensin II incited the interest of several research groups. This 3-8 fragment, denoted as Angiotensin IV (Ang IV) causes a number of distinct biological effects (see Introduction), unlikely to be explained by its weak binding to AT(1) and/or AT(2) receptors. Moreover the discovery of high affinity [(125)I]-Ang IV binding sites and their particular tissue distribution led to the concept of the AT(4) receptor. An important breakthrough was achieved by defining the AT(4) receptor as the membrane-bound insulin-regulated aminopeptidase (IRAP). Crucial for the definition as a receptor the binding of the endogenous ligand(s) should be linked to particular cellular and/or biochemical processes. With this respect, cultured cells offer the possibility to study the presence of binding sites in conjunction with ligand induced signaling. This link is discussed for the AT(4) receptor by providing an overview of the cellular effects by AT(4) ligands.
Collapse
Affiliation(s)
- Patrick M L Vanderheyden
- Department of Molecular and Biochemical Pharmacology, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
37
|
De Bundel D, Smolders I, Vanderheyden P, Michotte Y. Ang II and Ang IV: unraveling the mechanism of action on synaptic plasticity, memory, and epilepsy. CNS Neurosci Ther 2009; 14:315-39. [PMID: 19040556 DOI: 10.1111/j.1755-5949.2008.00057.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
The central angiotensin system plays a crucial role in cardiovascular regulation. More recently, angiotensin peptides have been implicated in stress, anxiety, depression, cognition, and epilepsy. Angiotensin II (Ang II) exerts its actions through AT(1) and AT(2) receptors, while most actions of its metabolite Ang IV were believed to be independent of AT(1) or AT(2) receptor activation. A specific binding site with high affinity for Ang IV was discovered and denominated "AT(4) receptor". The beneficiary effects of AT(4) ligands in animal models for cognitive impairment and epileptic seizures initiated the search for their mechanism of action. This proved to be a challenging task, and after 20 years of research, the nature of the "AT(4) receptor" remains controversial. Insulin-regulated aminopeptidase (IRAP) was first identified as the high-affinity binding site for AT(4) ligands. Recently, the hepatocyte growth factor receptor c-MET was also proposed as a receptor for AT(4) ligands. The present review focuses on the effects of Ang II and Ang IV on synaptic transmission and plasticity, learning, memory, and epileptic seizure activity. Possible interactions of Ang IV with the classical AT(1) and AT(2) receptor subtypes are evaluated, and other potential mechanisms by which AT(4) ligands may exert their effects are discussed. Identification of these mechanisms may provide a valuable target in the development in novel drugs for the treatment of cognitive disorders and epilepsy.
Collapse
Affiliation(s)
- Dimitri De Bundel
- Research Group Experimental Pharmacology, Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Vrije Universiteit Brussel, Brussels, Belgium
| | | | | | | |
Collapse
|
38
|
Chai SY, Yeatman HR, Parker MW, Ascher DB, Thompson PE, Mulvey HT, Albiston AL. Development of cognitive enhancers based on inhibition of insulin-regulated aminopeptidase. BMC Neurosci 2008; 9 Suppl 2:S14. [PMID: 19090987 PMCID: PMC2604898 DOI: 10.1186/1471-2202-9-s2-s14] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
The peptides angiotensin IV and LVV-hemorphin 7 were found to enhance memory in a number of memory tasks and reverse the performance deficits in animals with experimentally induced memory loss. These peptides bound specifically to the enzyme insulin-regulated aminopeptidase (IRAP), which is proposed to be the site in the brain that mediates the memory effects of these peptides. However, the mechanism of action is still unknown but may involve inhibition of the aminopeptidase activity of IRAP, since both angiotensin IV and LVV-hemorphin 7 are competitive inhibitors of the enzyme. IRAP also has another functional domain that is thought to regulate the trafficking of the insulin-responsive glucose transporter GLUT4, thereby influencing glucose uptake into cells. Although the exact mechanism by which the peptides enhance memory is yet to be elucidated, IRAP still represents a promising target for the development of a new class of cognitive enhancing agents.
Collapse
Affiliation(s)
- Siew Yeen Chai
- Howard Florey Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
- Centre for Neuroscience, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Holly R Yeatman
- Howard Florey Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
- Centre for Neuroscience, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Michael W Parker
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
- St Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia
| | - David B Ascher
- St Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia
| | - Philip E Thompson
- Department of Medicinal Chemistry, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Hayley T Mulvey
- Department of Medicinal Chemistry, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Anthony L Albiston
- Howard Florey Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
39
|
Abstract
Biology is replete with examples of hormesis, the term introduced and developed by Calabrese. The corresponding concept in the field of peptide research has been characterized as the inverted U-shaped dose-response relationship. The articles by Calabrese in this issue summarize the notable progress occurring in the past three decades. In contrast to the skepticism encountered when we introduced this concept for peptides in the early 1970s, hormesis is now becoming recognized as characteristic of many actions of these small proteins. Calabrese is performing a considerable service by his strong advocacy and promotion of the concept to a more general readership. Hopefully, hormesis will be routinely considered in the design of research projects and the discovery of pharmaceutical agents.
Collapse
Affiliation(s)
- Abba J Kastin
- Pennington Biomedical Research Center, Baton Rouge, Louisiana 70808, USA.
| | | |
Collapse
|
40
|
Albiston AL, Morton CJ, Ng HL, Pham V, Yeatman HR, Ye S, Fernando RN, De Bundel D, Ascher DB, Mendelsohn FAO, Parker MW, Chai SY. Identification and characterization of a new cognitive enhancer based on inhibition of insulin‐regulated aminopeptidase. FASEB J 2008; 22:4209-17. [DOI: 10.1096/fj.08-112227] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- Anthony L. Albiston
- Howard Florey Institute, Florey Neurosciences InstitutesParkvilleVictoriaAustralia
| | - Craig J. Morton
- St. Vincent's Institute of Medical ResearchFitzroyVictoriaAustralia
| | - Hooi Ling Ng
- St. Vincent's Institute of Medical ResearchFitzroyVictoriaAustralia
| | - Vi Pham
- Howard Florey Institute, Florey Neurosciences InstitutesParkvilleVictoriaAustralia
| | - Holly R. Yeatman
- Howard Florey Institute, Florey Neurosciences InstitutesParkvilleVictoriaAustralia
| | - Siying Ye
- Howard Florey Institute, Florey Neurosciences InstitutesParkvilleVictoriaAustralia
- Department of PhysiologyDartmouth Medical SchoolHanoverNHUSA
| | - Ruani N. Fernando
- Howard Florey Institute, Florey Neurosciences InstitutesParkvilleVictoriaAustralia
- Division of Molecular Neurobiology, Department of Medical Biochemistry and BiophysicsKarolinska InstituteStockholmSweden
| | - Dimitri De Bundel
- Howard Florey Institute, Florey Neurosciences InstitutesParkvilleVictoriaAustralia
- Research Group of Experimental Phar macology, Department of Pharmaceutical Chemistry, Drug Analysis and Drug InformationVrije UniversityBrusselBrusselsBelgium
| | - David B. Ascher
- St. Vincent's Institute of Medical ResearchFitzroyVictoriaAustralia
| | | | - Michael W. Parker
- Department of Biochemistry and Molecular BiologyBio21 Molecular Science and Biotechnology InstituteParkvilleVictoriaAustralia
- St. Vincent's Institute of Medical ResearchFitzroyVictoriaAustralia
| | - Siew Yeen Chai
- Howard Florey Institute, Florey Neurosciences InstitutesParkvilleVictoriaAustralia
- Centre for NeuroscienceUniversity of MelbourneParkvilleVictoriaAustralia
| |
Collapse
|
41
|
Wright JW, Yamamoto BJ, Harding JW. Angiotensin receptor subtype mediated physiologies and behaviors: new discoveries and clinical targets. Prog Neurobiol 2008; 84:157-81. [PMID: 18160199 PMCID: PMC2276843 DOI: 10.1016/j.pneurobio.2007.10.009] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2007] [Revised: 08/17/2007] [Accepted: 10/24/2007] [Indexed: 10/22/2022]
Abstract
The renin-angiotensin system (RAS) mediates several classic physiologies including body water and electrolyte homeostasis, blood pressure, cyclicity of reproductive hormones and sexual behaviors, and the regulation of pituitary gland hormones. These functions appear to be mediated by the angiotensin II (AngII)/AT(1) receptor subtype system. More recently, the angiotensin IV (AngIV)/AT(4) receptor subtype system has been implicated in cognitive processing, cerebroprotection, local blood flow, stress, anxiety and depression. There is accumulating evidence to suggest an inhibitory influence by AngII acting at the AT(1) subtype, and a facilitory role by AngIV acting at the AT(4) subtype, on neuronal firing rate, long-term potentiation, associative and spatial learning, and memory. This review initially describes the biochemical pathways that permit synthesis and degradation of active angiotensin peptides and three receptor subtypes (AT(1), AT(2) and AT(4)) thus far characterized. There is vigorous debate concerning the identity of the most recently discovered receptor subtype, AT(4). Descriptions of classic and novel physiologies and behaviors controlled by the RAS are presented. This review concludes with a consideration of the emerging therapeutic applications suggested by these newly discovered functions of the RAS.
Collapse
Affiliation(s)
- John W Wright
- Department of Psychology, Washington State University, P.O. Box 644820, Pullman, WA 99164-4820, USA.
| | | | | |
Collapse
|
42
|
Involvement of insulin-regulated aminopeptidase in the effects of the renin–angiotensin fragment angiotensin IV: a review. Heart Fail Rev 2007; 13:321-37. [DOI: 10.1007/s10741-007-9062-x] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2007] [Accepted: 10/16/2007] [Indexed: 10/22/2022]
|
43
|
Lanckmans K, Stragier B, Sarre S, Smolders I, Michotte Y. Nano-LC-MS/MS for the monitoring of angiotensin IV in rat brain microdialysates: Limitations and possibilities. J Sep Sci 2007; 30:2217-24. [PMID: 17688302 DOI: 10.1002/jssc.200700159] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
To broaden our knowledge about the central role of the angiotensin IV (Ang IV) peptide, we aimed to monitor its extracellular concentration in the brain using in vivo microdialysis. Ang IV was measured in the dialysates using a previously developed nano-LC-MS/MS assay with an LOD of 50 pM. Using this assay, baseline levels of Ang IV in dialysates from different brain structures were undetectable. However, immediately after microdialysis probe insertion, Ang IV could be detected in a concentration that varied between 120 and 187 pM. Using the zero-net-flux method, the extracellular levels of Ang IV in the striatum were estimated at 46 pM. These data may indicate that Ang IV is mainly present intracellularly. In addition, Ang IV was clearly measurable after striatal perfusion of Ang II. On the other hand, our nano-LC-MS/MS method was successful for the detection of Met-enkephalin and neurotensin in dialysates from the rat. In conclusion, the nano-LC-MS/MS method coupled with microdialysis is well suited to monitor the biologically significant conversion between Ang II and Ang IV in vivo, but physiological extracellular levels of Ang IV appear too low to be detected.
Collapse
Affiliation(s)
- Katrien Lanckmans
- Research Group Experimental Pharmacology, Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Vrije Universiteit Brussel, Brussels, Belgium
| | | | | | | | | |
Collapse
|
44
|
Albiston AL, Peck GR, Yeatman HR, Fernando R, Ye S, Chai SY. Therapeutic targeting of insulin-regulated aminopeptidase: heads and tails? Pharmacol Ther 2007; 116:417-27. [PMID: 17900701 DOI: 10.1016/j.pharmthera.2007.07.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2007] [Accepted: 07/30/2007] [Indexed: 10/22/2022]
Abstract
Insulin-regulated aminopeptidase, IRAP, is an abundant protein that was initially cloned from a rat epididymal fat pad cDNA library as a marker protein for specialized vesicles containing the insulin-responsive glucose transporter GLUT4, wherein it is thought to participate in the tethering and trafficking of GLUT4 vesicles. The same protein was independently cloned from human placental cDNA library as oxytocinase and is proposed to have a primary role in the regulation of circulating oxytocin (OXY) during the later stages of pregnancy. More recently, IRAP was identified as the specific binding site for angiotensin IV, and we propose that it mediates the memory-enhancing effects of the peptide. This protein appears to have multiple physiological roles that are tissue- and domain-specific; thus the protein can be specifically targeted for treating different clinical conditions.
Collapse
Affiliation(s)
- Anthony L Albiston
- Howard Florey Institute of Experimental Physiology and Medicine, University of Melbourne, Parkville, Victoria 3010, Australia
| | | | | | | | | | | |
Collapse
|
45
|
Fernando RN, Luff SE, Albiston AL, Chai SY. Sub-cellular localization of insulin-regulated membrane aminopeptidase, IRAP to vesicles in neurons. J Neurochem 2007; 102:967-76. [PMID: 17504262 DOI: 10.1111/j.1471-4159.2007.04659.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Angiotensin IV and LVV-hemorphin 7 promote robust enhancing effects on learning and memory. These peptides are also competitive inhibitors of the insulin-regulated membrane aminopeptidase, suggesting that the biological actions of these peptides may result from inhibition of IRAP activity. However, the normal function of IRAP in the brain is yet to be determined. The present study investigated the sub-cellular distribution of IRAP in four neuronal cell lines and in the mouse brain. Using sub-cellular fractionation, IRAP was found to be enriched in low density microsomes, while lower levels of IRAP were also present in high density microsomes, plasma membrane and mitochondrial fractions. Dual-label immunohistochemistry confirmed the presence of IRAP in vesicles co-localized with the vesicular maker VAMP2, in the trans Golgi network co-localized with TGN 38 and in endosomes co-localized with EEA1. Finally using electron microscopy, IRAP specific immunoreactivity was predominantly associated with large 100-200 nm vesicles in hippocampal neurons. The location, appearance and size of these vesicles are consistent with neurosecretory vesicles. IRAP precipitate was also detected in intracellular structures including the rough endoplasmic reticulum, Golgi stack and mitochondrial membranes. The sub-cellular localization of IRAP in neurons demonstrated in the present study bears striking parallels with distribution of IRAP in insulin responsive cells, where the enzyme plays a role in insulin-regulated glucose uptake. Therefore, we propose that the function of IRAP in neurons may be similar to that in insulin responsive cells.
Collapse
Affiliation(s)
- Ruani N Fernando
- Howard Florey Institute, The University of Melbourne, Parkville, Victoria, Australia
| | | | | | | |
Collapse
|
46
|
Mukuda T, Sugiyama H. An angiotensin II receptor antagonist suppresses running-enhanced hippocampal neurogenesis in rat. Neurosci Res 2007; 58:140-4. [PMID: 17397954 DOI: 10.1016/j.neures.2007.02.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2006] [Revised: 02/05/2007] [Accepted: 02/07/2007] [Indexed: 01/17/2023]
Abstract
Hippocampal neurogenesis is enhanced by voluntary running exercise in adult mammals. To elucidate the factors involved in this enhancement, we examined the effects of losartan, an antagonist of angiotensin II type 1 receptors, on the running-enhanced neurogenesis in the adult rat hippocampus. When losartan was administered orally via the drinking water, the running-enhanced cell proliferation in the subgranular zone was almost completely suppressed, indicating that this enhancement may be mediated by angiotensin II and its receptors.
Collapse
Affiliation(s)
- Takao Mukuda
- Department of Biology, Graduate School of Science, Kyushu University, 6-10-1 Hakozaki, Higashi-ku, Fukuoka 812-8581, Japan
| | | |
Collapse
|
47
|
Braszko JJ, Walesiuk A, Wielgat P. Cognitive effects attributed to angiotensin II may result from its conversion to angiotensin IV. J Renin Angiotensin Aldosterone Syst 2007; 7:168-74. [PMID: 17094054 DOI: 10.3317/jraas.2006.027] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
This study tests the hypothesis that the facilitation of learning and improvement of memory observed after an intracerebroventricular (i.c.v.) injection of angiotensin II (Ang II) is, in fact, caused by its derivative angiotensin IV (Ang IV). We ran two memory tests as well as an auxiliary test assessing motor performance in rats injected (i.c.v., 1 nmol in 2 microl saline) with Ang II or Ang IV. There were separate groups receiving peptide or saline five, 10 and 15 minutes before testing. Ang IV significantly increased step-through latencies in a passive avoidance paradigm as well as improved discrimination between familiar and unfamiliar objects in an object recognition test in all groups showing better retrieval of memory of aversive as well as appetitive stimuli in the peptide-treated groups regardless of the time of its injection. In contrast, rats treated with Ang II demonstrated significant improvement of memory of aversive and appetitive stimuli in the same tests only 15 minutes after its i.c.v. injection, with no effect in the groups injected five minutes before testing and slight efficacy in those injected 10 minutes before the test. Numbers of crossings, rearings and bar approaches in an open field were similar both in the peptide-treated and control groups making it unlikely that changes in motor performance affected the memory tests. In line with the present views on the intracellular metabolism of Ang II, these results suggest degradation to Ang IV by aminopeptidases A and N is necessary before the cognitive effects can occur.
Collapse
Affiliation(s)
- Jan Jozef Braszko
- Department of Clinical Pharmacology, Medical University of Bialystok, Bialystok, 15274, Poland.
| | | | | |
Collapse
|
48
|
Bonini JS, Bevilaqua LR, Zinn CG, Kerr DS, Medina JH, Izquierdo I, Cammarota M. Angiotensin II disrupts inhibitory avoidance memory retrieval. Horm Behav 2006; 50:308-13. [PMID: 16697382 DOI: 10.1016/j.yhbeh.2006.03.016] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2006] [Revised: 03/27/2006] [Accepted: 03/31/2006] [Indexed: 11/28/2022]
Abstract
The brain renin-angiotensin system (RAS) is involved in learning and memory, but the actual role of angiotensin II (A(II)) and its metabolites in this process has been difficult to comprehend. This has been so mainly due to procedural issues, especially the use of multi-trial learning paradigms and the utilization of pre-training intracerebroventricular infusion of RAS-acting compounds. Here, we specifically analyzed the action of A(II) in aversive memory retrieval using a hippocampal-dependent, one-trial, step-down inhibitory avoidance task (IA) in combination with stereotaxically localized intrahippocampal infusion of drugs. Rats bilaterally implanted with infusion cannulae aimed to the CA1 region of the dorsal hippocampus were trained in IA and tested for memory retention 24 h later. We found that when given into CA1 15 min before IA memory retention test, A(II), but not angiotensin IV or angiotensin(1-7) induced a dose-dependent and reversible amnesia without altering locomotor activity, exploratory behavior or anxiety state. The effect of A(II) was blocked in a dose-dependent manner by the A(II)-type 2 receptor (AT(2)) antagonist PD123319 but not by the A(II)-type 1 receptor (AT(1)) antagonist losartan. By themselves, neither PD123319 nor losartan had any effect on memory expression. Our data indicate that intra-CA1 A(II) hinders retrieval of avoidance memory through a process that involves activation of AT(2) receptors.
Collapse
Affiliation(s)
- Juliana S Bonini
- Centro de Memória, Instituto de Pesquisas Biomédicas, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | | | | | | | | | | | |
Collapse
|
49
|
von Bohlen und Halbach O, Albrecht D. The CNS renin-angiotensin system. Cell Tissue Res 2006; 326:599-616. [PMID: 16555051 DOI: 10.1007/s00441-006-0190-8] [Citation(s) in RCA: 178] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2006] [Accepted: 02/20/2006] [Indexed: 01/24/2023]
Abstract
The renin-angiotensin system (RAS) is one of the best-studied enzyme-neuropeptide systems in the brain and can serve as a model for the action of peptides on neuronal function in general. It is now well established that the brain has its own intrinsic RAS with all its components present in the central nervous system. The RAS generates a family of bioactive angiotensin peptides with variable biological and neurobiological activities. These include angiotensin-(1-8) [Ang II], angiotensin-(3-8) [Ang IV], and angiotensin-(1-7) [Ang-(1-7)]. These neuroactive forms of angiotensin act through specific receptors. Only Ang II acts through two different high-specific receptors, termed AT1 and AT2. Neuronal AT1 receptors mediate the stimulatory actions of Ang II on blood pressure, water and salt intake, and the secretion of vasopressin. In contrast, neuronal AT2 receptors have been implicated in the stimulation of apoptosis and as being antagonistic to AT1 receptors. Among the many potential effects mediated by stimulation of AT2 are neuronal regeneration after injury and the inhibition of pathological growth. Ang-(1-7) mediates its antihypertensive effects by stimulating the synthesis and release of vasodilator prostaglandins and nitric oxide and by potentiating the hypotensive effects of bradykinin. New data concerning the roles of Ang IV and Ang-(1-7) in cognition also support the existence of complex site-specific interactions between multiple angiotensins and multiple receptors in the mediation of important central functions of the RAS. Thus, the RAS of the brain is involved not only in the regulation of blood pressure, but also in the modulation of multiple additional functions in the brain, including processes of sensory information, learning, and memory, and the regulation of emotional responses.
Collapse
Affiliation(s)
- O von Bohlen und Halbach
- Interdisciplinary Center for Neurosciences (IZN), Department of Neuroanatomy, University of Heidelberg, Im Neuenheimer Feld 307, 69120 Heidelberg, Germany.
| | | |
Collapse
|
50
|
Braszko JJ. Valsartan abolishes most of the memory-improving effects of intracerebroventricular angiotensin II in rats. Clin Exp Hypertens 2006; 27:635-49. [PMID: 16303640 DOI: 10.1080/10641960500298723] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
UNLABELLED This study explores behavioral effects of angiotensin II (Ang II) and a potent AT(1) receptor inhibitor valsartan ((S)-N-valeryl-N-{[2'-(1H-tetrazol-5-yl)biphenyl-4-yl]-methyl}-valine). Male Wistar rats (160-180 g) were administered valsartan (10 mg/kg) orally followed, 2 hr later, by Ang II (1 nmol) given intracerebroventricularly (i.c.v., right lateral ventricle). Then 15 min later rats underwent behavioral testing: acquisition of conditioned avoidance responses (CARs), recall of a passive avoidance behavior, open field, elevated "plus" maze, and "chimney" test. Object recognition was tested 60 min after the i.c.v. injections. In addition, effect of valsartan on Ang II stimulated drinking of water was tested. We found that valsartan did not modify the Ang II facilitation of CARs acquisition but abolished the Ang II improvement of memory retrieval and consolidation. The lack of effect of our treatments on the rats' motor activity in the open field makes unspecific contribution of the drug-induced performance changes to the cognitive tests improbable. The anxiogenic action of Ang II, decreased by valsartan, makes an unspecific influence of anxiety possible. The prevention of Ang II increase of drinking by orally given valsartan confirmed effective blockade of the brain AT(1) receptors by the drug. IN CONCLUSION valsartan appears to affect cognitive effects of i.c.v. Ang II in rats in a similar way to losartan; anxiolytic activity of valsartan appears to be slightly weaker than that of losartan.
Collapse
Affiliation(s)
- Jan J Braszko
- Department of Clinical Pharmacology, Medical Academy of Bialystok, Bialystok, Poland.
| |
Collapse
|