1
|
Caniceiro AB, Bueschbell B, Schiedel AC, Moreira IS. Class A and C GPCR Dimers in Neurodegenerative Diseases. Curr Neuropharmacol 2022; 20:2081-2141. [PMID: 35339177 PMCID: PMC9886835 DOI: 10.2174/1570159x20666220327221830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 02/21/2022] [Accepted: 03/23/2022] [Indexed: 11/22/2022] Open
Abstract
Neurodegenerative diseases affect over 30 million people worldwide with an ascending trend. Most individuals suffering from these irreversible brain damages belong to the elderly population, with onset between 50 and 60 years. Although the pathophysiology of such diseases is partially known, it remains unclear upon which point a disease turns degenerative. Moreover, current therapeutics can treat some of the symptoms but often have severe side effects and become less effective in long-term treatment. For many neurodegenerative diseases, the involvement of G proteincoupled receptors (GPCRs), which are key players of neuronal transmission and plasticity, has become clearer and holds great promise in elucidating their biological mechanism. With this review, we introduce and summarize class A and class C GPCRs, known to form heterodimers or oligomers to increase their signalling repertoire. Additionally, the examples discussed here were shown to display relevant alterations in brain signalling and had already been associated with the pathophysiology of certain neurodegenerative diseases. Lastly, we classified the heterodimers into two categories of crosstalk, positive or negative, for which there is known evidence.
Collapse
Affiliation(s)
- Ana B. Caniceiro
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; ,These authors contributed equally to this work.
| | - Beatriz Bueschbell
- PhD Programme in Experimental Biology and Biomedicine, Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Casa Costa Alemão, 3030-789 Coimbra, Portugal; ,These authors contributed equally to this work.
| | - Anke C. Schiedel
- Department of Pharmaceutical & Medicinal Chemistry, Pharmaceutical Institute, University of Bonn, D-53121 Bonn, Germany;
| | - Irina S. Moreira
- University of Coimbra, Department of Life Sciences, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal; ,Center for Neuroscience and Cell Biology, Center for Innovative Biomedicine and Biotechnology, 3004-504 Coimbra, Portugal,Address correspondence to this author at the Center for Neuroscience and Cell Biology, Center for Innovative Biomedicine and Biotechnology, 3004-504 Coimbra, Portugal; E-mail:
| |
Collapse
|
2
|
Grotemeyer A, McFleder RL, Wu J, Wischhusen J, Ip CW. Neuroinflammation in Parkinson's Disease - Putative Pathomechanisms and Targets for Disease-Modification. Front Immunol 2022; 13:878771. [PMID: 35663989 PMCID: PMC9158130 DOI: 10.3389/fimmu.2022.878771] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 04/20/2022] [Indexed: 12/15/2022] Open
Abstract
Parkinson’s disease (PD) is a progressive and debilitating chronic disease that affects more than six million people worldwide, with rising prevalence. The hallmarks of PD are motor deficits, the spreading of pathological α-synuclein clusters in the central nervous system, and neuroinflammatory processes. PD is treated symptomatically, as no causally-acting drug or procedure has been successfully established for clinical use. Various pathways contributing to dopaminergic neuron loss in PD have been investigated and described to interact with the innate and adaptive immune system. We discuss the possible contribution of interconnected pathways related to the immune response, focusing on the pathophysiology and neurodegeneration of PD. In addition, we provide an overview of clinical trials targeting neuroinflammation in PD.
Collapse
Affiliation(s)
| | | | - Jingjing Wu
- Department of Neurology, University Hospital Würzburg, Würzburg, Germany
| | - Jörg Wischhusen
- Section for Experimental Tumor Immunology, Department of Obstetrics and Gynecology, University Hospital of Würzburg, Würzburg, Germany
| | - Chi Wang Ip
- Department of Neurology, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
3
|
AngIV-Analog Dihexa Rescues Cognitive Impairment and Recovers Memory in the APP/PS1 Mouse via the PI3K/AKT Signaling Pathway. Brain Sci 2021; 11:brainsci11111487. [PMID: 34827486 PMCID: PMC8615599 DOI: 10.3390/brainsci11111487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 10/29/2021] [Accepted: 11/09/2021] [Indexed: 11/17/2022] Open
Abstract
The renin-angiotensin system (RAS) is a paracrine RAS within the central nervous system (CNS) and is closely related to Alzheimer’s disease (AD). The endogenous hexapeptide angiotensin IV (Ang IV), an important component of the brain RAS, was found to rescue cognitive impairment and recover memory in previous studies. In our study, we used different doses of Dihexa, which can be orally administered and cross the BBB in APP/PS1 mice. We found that the amount of AngIV in mouse tissue increased after the administration of Dihexa compared to that in the WT group. Meanwhile, Dihexa restored spatial learning and cognitive functions in the Morris water maze test. Dihexa increased the neuronal cells and the expression of SYP protein in APP/PS1 mice in Nissl staining. Furthermore, Dihexa decreased the activation of astrocytes and microglia, markedly reduced levels of the pro-inflammatory cytokines IL-1β and TNF-α and increased the levels of the anti-inflammatory cytokine IL-10. Dihexa activated the PI3K/AKT signaling pathway, while PI3K inhibitor wortmannin significantly reversed the anti-inflammatory and anti-apoptotic effects of APP/PS1 mice. These findings highlight the brain AngIV/PI3K/AKT axis as a potential target for the treatment of AD.
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: 2.4] [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
|
Hallberg M, Larhed M. From Angiotensin IV to Small Peptidemimetics Inhibiting Insulin-Regulated Aminopeptidase. Front Pharmacol 2020; 11:590855. [PMID: 33178027 PMCID: PMC7593869 DOI: 10.3389/fphar.2020.590855] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 09/18/2020] [Indexed: 12/26/2022] Open
Abstract
It was reported three decades ago that intracerebroventricular injection of angiotensin IV (Ang IV, Val-Tyr-Ile-His-Pro-Phe) improved memory and learning in the rat. There are several explanations for these positive effects of the hexapeptide and related analogues on cognition available in the literature. In 2001, it was proposed that the insulin-regulated aminopeptidase (IRAP) is a main target for Ang IV and that Ang IV serves as an inhibitor of the enzyme. The focus of this review is the efforts to stepwise transform the hexapeptide into more drug-like Ang IV peptidemimetics serving as IRAP inhibitors. Moreover, the discovery of IRAP inhibitors by virtual and substance library screening and direct design applying knowledge of the structure of IRAP and of related enzymes is briefly presented.
Collapse
Affiliation(s)
- Mathias Hallberg
- The Beijer Laboratory, Division of Biological Research on Drug Dependence, Department of Pharmaceutical Biosciences, BMC, Uppsala University, Uppsala, Sweden
| | - Mats Larhed
- Department of Medicinal Chemistry, Science for Life Laboratory, BMC, Uppsala University, Uppsala, Sweden
| |
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: 28] [Impact Index Per Article: 5.6] [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
|
Wright JW, Harding JW. Contributions by the Brain Renin-Angiotensin System to Memory, Cognition, and Alzheimer's Disease. J Alzheimers Dis 2020; 67:469-480. [PMID: 30664507 DOI: 10.3233/jad-181035] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by progressive neuron losses in memory-associated brain structures that rob patients of their dignity and quality of life. Five drugs have been approved by the FDA to treat AD but none modify or significantly slow disease progression. New therapies are needed to delay the course of this disease with the ultimate goal of preventing neuron losses and preserving memory functioning. In this review we describe the renin-angiotensin II (AngII) system (RAS) with specific regard to its deleterious contributions to hypertension, facilitation of neuroinflammation and oxidative stress, reduced cerebral blood flow, tissue remodeling, and disruption of memory consolidation and retrieval. There is evidence that components of the RAS, AngIV and Ang(1-7), are positioned to counter such damaging influences and these systems are detailed with the goal of drawing attention to their importance as drug development targets. Ang(1-7) binds at the Mas receptor, while AngIV binds at the AT4 receptor subtype, and these receptor numbers are significantly decreased in AD patients, accompanied by declines in brain aminopeptidases A and N, enzymes essential for the synthesis of AngIV. Potent analogs may be useful to counter these changes and facilitate neuronal functioning and reduce apoptosis in memory associated brain structures of AD patients.
Collapse
Affiliation(s)
- John W Wright
- Department of Psychology, Washington State University, Pullman, WA, USA.,Department of Integrative Physiology and Neuroscience, and Program in Biotechnology, Washington State University, Pullman, WA, USA.,M3 Biotechnology, Inc., Seattle, WA, USA
| | - Joseph W Harding
- Department of Psychology, Washington State University, Pullman, WA, USA.,Department of Integrative Physiology and Neuroscience, and Program in Biotechnology, Washington State University, Pullman, WA, USA.,M3 Biotechnology, Inc., Seattle, WA, USA
| |
Collapse
|
8
|
Evans CE, Miners JS, Piva G, Willis CL, Heard DM, Kidd EJ, Good MA, Kehoe PG. ACE2 activation protects against cognitive decline and reduces amyloid pathology in the Tg2576 mouse model of Alzheimer's disease. Acta Neuropathol 2020; 139:485-502. [PMID: 31982938 PMCID: PMC7035243 DOI: 10.1007/s00401-019-02098-6] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 10/16/2019] [Accepted: 11/08/2019] [Indexed: 02/07/2023]
Abstract
Mid-life hypertension and cerebrovascular dysfunction are associated with increased risk of later life dementia, including Alzheimer’s disease (AD). The classical renin–angiotensin system (cRAS), a physiological regulator of blood pressure, functions independently within the brain and is overactive in AD. cRAS-targeting anti-hypertensive drugs are associated with reduced incidence of AD, delayed onset of cognitive decline, and reduced levels of Aβ and tau in both animal models and human pathological studies. cRAS activity is moderated by a downstream regulatory RAS pathway (rRAS), which is underactive in AD and is strongly associated with pathological hallmarks in human AD, and cognitive decline in animal models of CNS disease. We now show that enhancement of brain ACE2 activity, a major effector of rRAS, by intraperitoneal administration of diminazene aceturate (DIZE), an established activator of ACE2, lowered hippocampal Aβ and restored cognition in mid-aged (13–14-month-old) symptomatic Tg2576 mice. We confirmed that the protective effects of DIZE were directly mediated through ACE2 and were associated with reduced hippocampal soluble Aβ42 and IL1-β levels. DIZE restored hippocampal MasR levels in conjunction with increased NMDA NR2B and downstream ERK signalling expression in hippocampal synaptosomes from Tg2576 mice. Chronic (10 weeks) administration of DIZE to pre-symptomatic 9–10-month-old Tg2576 mice, and acute (10 days) treatment in cognitively impaired 12–13-month-old mice, prevented the development of cognitive impairment. Together these data demonstrate that ACE2 enhancement protects against and reverses amyloid-related hippocampal pathology and cognitive impairment in a preclinical model of AD.
Collapse
|
9
|
Seyer B, Diwakarla S, Burns P, Hallberg A, Grӧnbladh A, Hallberg M, Chai SY. Insulin-regulated aminopeptidase inhibitor-mediated increases in dendritic spine density are facilitated by glucose uptake. J Neurochem 2019; 153:485-494. [PMID: 31556456 DOI: 10.1111/jnc.14880] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 09/08/2019] [Accepted: 09/11/2019] [Indexed: 02/06/2023]
Abstract
Ethyl2-acetylamino-7-hydroxy-4-pyridin-3-yl-4H-chromene-3-carboxylate (HFI-419), the benzopyran-based inhibitor of insulin-regulated aminopeptidase (IRAP), has previously been shown to improve spatial working and recognition memory in rodents. However, the mechanism of its cognitive-enhancing effect remains unknown. There is a close correlation between dendritic spine density and learning in vivo and several studies suggest that increases in neuronal glucose uptake and/or alterations to the activity of matrix metalloproteinases (MMPs) may improve memory and increase dendritic spine density. We aimed to identify the potential mechanism by which HFI-419 enhances memory by utilizing rat primary cultures of hippocampal cells. Alterations to dendritic spine density were assessed in the presence of varying concentrations of HFI-419 at different stages of hippocampal cell development. In addition, glucose uptake and changes to spine density were assessed in the presence of indinavir, an inhibitor of the glucose transporter 4 (GLUT4 ), or the matrix metalloprotease inhibitor CAS 204140-01-2. We confirmed that inhibition of IRAP activity with HFI-419 enhanced spatial working memory in rats, and determined that this enhancement may be driven by GLUT4 -mediated changes to dendritic spine density. We observed that IRAP inhibition increased dendritic spine density prior to peak dendritic growth in hippocampal neurons, and that spine formation was inhibited when GLUT4 -mediated glucose uptake was blocked. In addition, during the peak phase of dendritic spine growth, the effect of IRAP inhibition on enhancement of dendritic spine density resulted specifically in an increase in the proportion of mushroom/stubby-like spines, a morphology associated with memory and learning. Moreover, these spines were deemed to be functional based on their expression of the pre-synaptic markers vesicular glutamate transporter 1 and synapsin. Overall, or findings suggest that IRAP inhibitors may facilitate memory by increasing hippocampal dendritic spine density via a GLUT4 -mediated mechanism. Cover Image for this issue: doi: 10.1111/jnc.14745.
Collapse
Affiliation(s)
- Benjamin Seyer
- Faculty of Biomedical and Psychological Sciences, Department of Physiology, Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Shanti Diwakarla
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence, Uppsala University, BMC, Uppsala, Sweden
| | - Peta Burns
- Faculty of Biomedical and Psychological Sciences, Department of Physiology, Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Anders Hallberg
- Department of Medicinal Chemistry, Uppsala University, BMC, Uppsala, Sweden
| | - Alfhild Grӧnbladh
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence, Uppsala University, BMC, Uppsala, Sweden
| | - Mathias Hallberg
- The Beijer Laboratory, Department of Pharmaceutical Biosciences, Division of Biological Research on Drug Dependence, Uppsala University, BMC, Uppsala, Sweden
| | - Siew Yeen Chai
- Faculty of Biomedical and Psychological Sciences, Department of Physiology, Biomedicine Discovery Institute, Monash University, Clayton, Australia
| |
Collapse
|
10
|
Wang QG, Xue X, Yang Y, Gong PY, Jiang T, Zhang YD. Angiotensin IV suppresses inflammation in the brains of rats with chronic cerebral hypoperfusion. J Renin Angiotensin Aldosterone Syst 2019; 19:1470320318799587. [PMID: 30223703 PMCID: PMC6144503 DOI: 10.1177/1470320318799587] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
INTRODUCTION This study aimed to evaluate the influence of central angiotensin IV (Ang IV) infusion on chronic cerebral hypoperfusion (CCH)-related neuropathological changes including amyloid-β (Aβ), hyperphosphorylated tau (p-tau) and the inflammatory response. MATERIALS AND METHODS Rats with CCH received central infusion of Ang IV, its receptor AT4R antagonist divalinal-Ang IV or artificial cerebrospinal fluid for six weeks. During this procedure, the systolic blood pressure (SBP) was monitored, and the levels of Aβ42, p-tau and pro-inflammatory cytokines in the brain were detected. RESULTS Rats with CCH exhibited higher levels of Aβ42, p-tau and pro-inflammatory cytokines in the brain when compared with controls. Infusion of Ang IV significantly reduced the expression of pro-inflammatory cytokines in the brains of rats with CCH. Meanwhile, the reduction of pro-inflammatory cytokines levels caused by Ang IV was reversed by divalinal-Ang IV. During the treatment, the SBP in rats was not significantly altered. CONCLUSION This study demonstrates for the first time that Ang IV dose-dependently suppresses inflammation through AT4R in the brains of rats with CCH, which is independent from SBP. These findings suggest that Ang IV/AT4R may represent a potential therapeutic target for CCH-related neurological diseases.
Collapse
Affiliation(s)
- Qing-Guang Wang
- 1 Department of Neurology, Nanjing First Hospital, Nanjing Medical University, People's Republic of China.,2 Department of Neurology, Jiangyin People's Hospital, Nanjing Medical University, People's Republic of China
| | - Xiao Xue
- 1 Department of Neurology, Nanjing First Hospital, Nanjing Medical University, People's Republic of China
| | - Yang Yang
- 1 Department of Neurology, Nanjing First Hospital, Nanjing Medical University, People's Republic of China
| | - Peng-Yu Gong
- 1 Department of Neurology, Nanjing First Hospital, Nanjing Medical University, People's Republic of China
| | - Teng Jiang
- 1 Department of Neurology, Nanjing First Hospital, Nanjing Medical University, People's Republic of China
| | - Ying-Dong Zhang
- 1 Department of Neurology, Nanjing First Hospital, Nanjing Medical University, People's Republic of China
| |
Collapse
|
11
|
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: 37] [Impact Index Per Article: 5.3] [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
|
12
|
Vanga SR, Sävmarker J, Ng L, Larhed M, Hallberg M, Åqvist J, Hallberg A, Chai SY, Gutiérrez-de-Terán H. Structural Basis of Inhibition of Human Insulin-Regulated Aminopeptidase (IRAP) by Aryl Sulfonamides. ACS OMEGA 2018; 3:4509-4521. [PMID: 30023895 PMCID: PMC6045421 DOI: 10.1021/acsomega.8b00595] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 04/16/2018] [Indexed: 05/07/2023]
Abstract
The insulin-regulated aminopeptidase (IRAP) is a membrane-bound zinc metallopeptidase with many important regulatory functions. It has been demonstrated that inhibition of IRAP by angiotensin IV (Ang IV) and other peptides, as well as more druglike inhibitors, improves cognition in several rodent models. We recently reported a series of aryl sulfonamides as small-molecule IRAP inhibitors and a promising scaffold for pharmacological intervention. We have now expanded with a number of derivatives, report their stability in liver microsomes, and characterize the activity of the whole series in a new assay performed on recombinant human IRAP. Several compounds, such as the new fluorinated derivative 29, present submicromolar affinity and high metabolic stability. Starting from the two binding modes previously proposed for the sulfonamide scaffold, we systematically performed molecular dynamics simulations and binding affinity estimation with the linear interaction energy method for the full compound series. The significant agreement with experimental affinities suggests one of the binding modes, which was further confirmed by the excellent correlation for binding affinity differences between the selected pair of compounds obtained by rigorous free energy perturbation calculations. The new experimental data and the computationally derived structure-activity relationship of the sulfonamide series provide valuable information for further lead optimization of novel IRAP inhibitors.
Collapse
Affiliation(s)
- Sudarsana Reddy Vanga
- Department
of Cell and Molecular Biology, BMC, Box 596, Uppsala University, SE-751
24 Uppsala, Sweden
| | - Jonas Sävmarker
- Department of Medicinal Chemistry and Science for Life Laboratory, Department
of Medicinal Chemistry, Uppsala University,
BMC, SE-751 24 Uppsala, Sweden
| | - Leelee Ng
- Biomedicine
Discovery Institute, Department of Physiology, Monash University, Clayton, Victoria 3800, Australia
| | - Mats Larhed
- Department of Medicinal Chemistry and Science for Life Laboratory, Department
of Medicinal Chemistry, Uppsala University,
BMC, SE-751 24 Uppsala, Sweden
| | - Mathias Hallberg
- The
Beijer Laboratory, Department of Pharmaceutical Biosciences, Division
of Biological Research on Drug Dependence, Uppsala University, BMC, SE-751 23 Uppsala, Sweden
| | - Johan Åqvist
- Department
of Cell and Molecular Biology, BMC, Box 596, Uppsala University, SE-751
24 Uppsala, Sweden
| | - Anders Hallberg
- Department of Medicinal Chemistry and Science for Life Laboratory, Department
of Medicinal Chemistry, Uppsala University,
BMC, SE-751 24 Uppsala, Sweden
| | - Siew Yeen Chai
- Biomedicine
Discovery Institute, Department of Physiology, Monash University, Clayton, Victoria 3800, Australia
- E-mail: . Phone: +61 3 990 52515. Fax: +61 3 990 52547 (S.Y.C.)
| | - Hugo Gutiérrez-de-Terán
- Department
of Cell and Molecular Biology, BMC, Box 596, Uppsala University, SE-751
24 Uppsala, Sweden
- E-mail: . Phone: +46 18 471 5056. Fax: +46 18 53 69 71 (H.G.-d.-T.)
| |
Collapse
|
13
|
Papadopoulos P, Tong XK, Imboden H, Hamel E. Losartan improves cerebrovascular function in a mouse model of Alzheimer's disease with combined overproduction of amyloid-β and transforming growth factor-β1. J Cereb Blood Flow Metab 2017; 37:1959-1970. [PMID: 27389178 PMCID: PMC5464692 DOI: 10.1177/0271678x16658489] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Alterations of the renin-angiotensin system have been implicated in the pathogenesis of Alzheimer's disease. We tested the efficacy of losartan (10 mg/kg/day for three months), a selective angiotensin II type 1 receptor antagonist, in alleviating cerebrovascular and cognitive deficits in double-transgenic mice (six months at endpoint) that overexpress a mutated form of the human amyloid precursor protein (APPSwe,Ind) and a constitutively active form of the transforming growth factor-β1, thereafter named A/T mice. Losartan rescued cerebrovascular reactivity, particularly the dilatory responses, but failed to attenuate astroglial activation and to normalize the neurovascular uncoupling response to sensory stimulation. The cognitive deficits of A/T mice were not restored by losartan nor were the increased brain levels of soluble and insoluble Aβ1-40 and Aβ1-42 peptides normalized. Our results are the first to demonstrate the capacity of losartan to improve cerebrovascular reactivity in an Alzheimer's disease mouse model of combined Aβ-induced vascular oxidative stress and transforming growth factor-β1-mediated vascular fibrosis. These data suggest that losartan may be promising for restoring cerebrovascular function in patients with vascular diseases at risk for vascular dementia or Alzheimer's disease. However, a combined therapy may be warranted for rescuing both vascular and cognitive deficits in a multifaceted pathology like Alzheimer's disease.
Collapse
Affiliation(s)
- Panayiota Papadopoulos
- 1 Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, Québec, Canada
| | - Xin-Kang Tong
- 1 Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, Québec, Canada
| | - Hans Imboden
- 2 Institute of Cell Biology, University of Bern, Bern, Switzerland
| | - Edith Hamel
- 1 Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, Québec, Canada
| |
Collapse
|
14
|
Mascolo A, Sessa M, Scavone C, De Angelis A, Vitale C, Berrino L, Rossi F, Rosano G, Capuano A. New and old roles of the peripheral and brain renin-angiotensin-aldosterone system (RAAS): Focus on cardiovascular and neurological diseases. Int J Cardiol 2016; 227:734-742. [PMID: 27823897 DOI: 10.1016/j.ijcard.2016.10.069] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 10/26/2016] [Indexed: 02/06/2023]
Abstract
It is commonly accepted that the renin-angiotensin-aldosterone system (RAAS) is a cardiovascular circulating hormonal system that plays also an important role in the modulation of several patterns in the brain. The pathway of the RAAS can be divided into two classes: the traditional pathway of RAAS, also named classic RAAS, and the non-classic RAAS. Both pathways play a role in both cardiovascular and neurological diseases through a peripheral or central control. In this regard, renewed interest is growing in the last years for the consideration that the brain RAAS could represent a new important therapeutic target to regulate not only the blood pressure via central nervous control, but also neurological diseases. However, the development of compounds able to cross the blood-brain barrier and to act on the brain RAAS is challenging, especially if the metabolic stability and the half-life are taken into consideration. To date, two drug classes (aminopeptidase type A inhibitors and angiotensin IV analogues) acting on the brain RAAS are in development in pre-clinical or clinical stages. In this article, we will present an overview of the biological functions played by peripheral and brain classic and non-classic pathways of the RAAS in several clinical conditions, focusing on the brain RAAS and on the new pharmacological targets of the RAAS.
Collapse
Affiliation(s)
- A Mascolo
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Second University of Naples, Naples, Italy.
| | - M Sessa
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Second University of Naples, Naples, Italy
| | - C Scavone
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Second University of Naples, Naples, Italy
| | - A De Angelis
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Second University of Naples, Naples, Italy
| | - C Vitale
- IRCCS San Raffaele Pisana, Rome, Italy
| | - L Berrino
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Second University of Naples, Naples, Italy
| | - F Rossi
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Second University of Naples, Naples, Italy
| | - G Rosano
- IRCCS San Raffaele Pisana, Rome, Italy; Cardiovascular and Cell Sciences Research Institute, St. George's, University of London, London, UK
| | - A Capuano
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Second University of Naples, Naples, Italy
| |
Collapse
|
15
|
Diwakarla S, Nylander E, Grönbladh A, Vanga SR, Shamsudin Y, Gutiérrez-de-Terán H, Sävmarker J, Ng L, Pham V, Lundbäck T, Jenmalm-Jensen A, Svensson R, Artursson P, Zelleroth S, Engen K, Rosenström U, Larhed M, Åqvist J, Chai SY, Hallberg M. Aryl Sulfonamide Inhibitors of Insulin-Regulated Aminopeptidase Enhance Spine Density in Primary Hippocampal Neuron Cultures. ACS Chem Neurosci 2016; 7:1383-1392. [PMID: 27501164 DOI: 10.1021/acschemneuro.6b00146] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The zinc metallopeptidase insulin regulated aminopeptidase (IRAP), which is highly expressed in the hippocampus and other brain regions associated with cognitive function, has been identified as a high-affinity binding site of the hexapeptide angiotensin IV (Ang IV). This hexapeptide is thought to facilitate learning and memory by binding to the catalytic site of IRAP to inhibit its enzymatic activity. In support of this hypothesis, low molecular weight, nonpeptide specific inhibitors of IRAP have been shown to enhance memory in rodent models. Recently, it was demonstrated that linear and macrocyclic Ang IV-derived peptides can alter the shape and increase the number of dendritic spines in hippocampal cultures, properties associated with enhanced cognitive performance. After screening a library of 10 500 drug-like substances for their ability to inhibit IRAP, we identified a series of low molecular weight aryl sulfonamides, which exhibit no structural similarity to Ang IV, as moderately potent IRAP inhibitors. A structural and biological characterization of three of these aryl sulfonamides was performed. Their binding modes to human IRAP were explored by docking calculations combined with molecular dynamics simulations and binding affinity estimations using the linear interaction energy method. Two alternative binding modes emerged from this analysis, both of which correctly rank the ligands according to their experimental binding affinities for this series of compounds. Finally, we show that two of these drug-like IRAP inhibitors can alter dendritic spine morphology and increase spine density in primary cultures of hippocampal neurons.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Leelee Ng
- Biomedicine Discovery Institute, Department of Physiology, Monash University , Clayton, Victoria 3800, Australia
| | - Vi Pham
- Biomedicine Discovery Institute, Department of Physiology, Monash University , Clayton, Victoria 3800, Australia
| | - Thomas Lundbäck
- Chemical Biology Consortium Sweden, Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medicinal Biochemistry and Biophysics, Karolinska Institute , 171 77 Solna, Sweden
| | - Annika Jenmalm-Jensen
- Chemical Biology Consortium Sweden, Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medicinal Biochemistry and Biophysics, Karolinska Institute , 171 77 Solna, Sweden
| | | | | | | | | | | | | | | | - Siew Yeen Chai
- Biomedicine Discovery Institute, Department of Physiology, Monash University , Clayton, Victoria 3800, Australia
| | | |
Collapse
|
16
|
Overview of Antagonists Used for Determining the Mechanisms of Action Employed by Potential Vasodilators with Their Suggested Signaling Pathways. Molecules 2016; 21:495. [PMID: 27092479 PMCID: PMC6274436 DOI: 10.3390/molecules21040495] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 03/23/2016] [Accepted: 03/28/2016] [Indexed: 01/04/2023] Open
Abstract
This paper is a review on the types of antagonists and the signaling mechanism pathways that have been used to determine the mechanisms of action employed for vasodilation by test compounds. Thus, we exhaustively reviewed and analyzed reports related to this topic published in PubMed between the years of 2010 till 2015. The aim of this paperis to suggest the most appropriate type of antagonists that correspond to receptors that would be involved during the mechanistic studies, as well as the latest signaling pathways trends that are being studied in order to determine the route(s) that atest compound employs for inducing vasodilation. The methods to perform the mechanism studies were included. Fundamentally, the affinity, specificity and selectivity of the antagonists to their receptors or enzymes were clearly elaborated as well as the solubility and reversibility. All the signaling pathways on the mechanisms of action involved in the vascular tone regulation have been well described in previous review articles. However, the most appropriate antagonists that should be utilized have never been suggested and elaborated before, hence the reason for this review.
Collapse
|
17
|
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: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [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
|
18
|
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 PMCID: PMC11482419 DOI: 10.1007/s10571-015-0285-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [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
|
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: 35] [Impact Index Per Article: 3.2] [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 PMCID: PMC4201273 DOI: 10.1124/jpet.114.218735] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 09/02/2014] [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: 9.8] [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
|
The environmental neurotoxicant PCB 95 promotes synaptogenesis via ryanodine receptor-dependent miR132 upregulation. J Neurosci 2014; 34:717-25. [PMID: 24431430 DOI: 10.1523/jneurosci.2884-13.2014] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Non-dioxin-like (NDL) polychlorinated biphenyls (PCBs) are widespread environmental contaminants linked to neuropsychological dysfunction in children. NDL PCBs increase spontaneous Ca(2+) oscillations in neurons by stabilizing ryanodine receptor (RyR) calcium release channels in the open configuration, which results in CREB-dependent dendritic outgrowth. In this study, we address the question of whether activation of CREB by NDL PCBs also triggers dendritic spine formation. Nanomolar concentrations of PCB 95, a NDL congener with potent RyR activity, significantly increased spine density and the frequency of miniature EPSCs in primary dissociated rat hippocampal cultures coincident with upregulation of miR132. Inhibition of RyR, CREB, or miR132 as well as expression of a mutant p250GAP cDNA construct that is not suppressed by miR132 blocked PCB 95 effects on spines and miniature EPSCs. PCB 95 also induced spine formation via RyR- and miR132-dependent mechanisms in hippocampal slice cultures. These data demonstrate a novel mechanism of PCB developmental neurotoxicity whereby RyR sensitization modulates spine formation and synaptogenesis via CREB-mediated miR132 upregulation, which in turn suppresses the translation of p250GAP, a negative regulator of synaptogenesis. In light of recent evidence implicating miR132 dysregulation in Rett syndrome and schizophrenia, these findings identify NDL PCBs as potential environmental risk factors for neurodevelopmental disorders.
Collapse
|
23
|
Wright JW, Kawas LH, Harding JW. A Role for the Brain RAS in Alzheimer's and Parkinson's Diseases. Front Endocrinol (Lausanne) 2013; 4:158. [PMID: 24298267 PMCID: PMC3829467 DOI: 10.3389/fendo.2013.00158] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Accepted: 10/09/2013] [Indexed: 12/30/2022] Open
Abstract
The brain renin-angiotensin system (RAS) has available the necessary functional components to produce the active ligands angiotensins II (AngII), angiotensin III, angiotensins (IV), angiotensin (1-7), and angiotensin (3-7). These ligands interact with several receptor proteins including AT1, AT2, AT4, and Mas distributed within the central and peripheral nervous systems as well as local RASs in several organs. This review first describes the enzymatic pathways in place to synthesize these ligands and the binding characteristics of these angiotensin receptor subtypes. We next discuss current hypotheses to explain the disorders of Alzheimer's disease (AD) and Parkinson's disease (PD), as well as research efforts focused on the use of angiotensin converting enzyme (ACE) inhibitors and angiotensin receptor blockers (ARBs), in their treatment. ACE inhibitors and ARBs are showing promise in the treatment of several neurodegenerative pathologies; however, there is a need for the development of analogs capable of penetrating the blood-brain barrier and acting as agonists or antagonists at these receptor sites. AngII and AngIV have been shown to play opposing roles regarding memory acquisition and consolidation in animal models. We discuss the development of efficacious AngIV analogs in the treatment of animal models of AD and PD. These AngIV analogs act via the AT4 receptor subtype which may coincide with the hepatocyte growth factor/c-Met receptor system. Finally, future research directions are described concerning new approaches to the treatment of these two neurological diseases.
Collapse
Affiliation(s)
- John W. Wright
- Departments of Psychology, Integrative Physiology and Neuroscience, Program in Biotechnology, Washington State University, Pullman, WA, USA
| | - Leen H. Kawas
- Departments of Psychology, Integrative Physiology and Neuroscience, Program in Biotechnology, Washington State University, Pullman, WA, USA
| | - Joseph W. Harding
- Departments of Psychology, Integrative Physiology and Neuroscience, Program in Biotechnology, Washington State University, Pullman, WA, USA
| |
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: 1.7] [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
|
McCoy AT, Benoist CC, Wright JW, Kawas LH, Bule-Ghogare JM, Zhu M, Appleyard SM, Wayman GA, Harding JW. Evaluation of metabolically stabilized angiotensin IV analogs as procognitive/antidementia agents. J Pharmacol Exp Ther 2013; 344:141-54. [PMID: 23055539 PMCID: PMC3533412 DOI: 10.1124/jpet.112.199497] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Accepted: 10/09/2012] [Indexed: 12/30/2022] Open
Abstract
Angiotensin IV (AngIV: VYIHPF)-related peptides have long been recognized as procognitive agents with potential as antidementia therapeutics. Their development as useful therapeutics, however, has been limited by physiochemical properties that make them susceptible to metabolic degradation and impermeable to gut and blood-brain barriers. A previous study demonstrated that the core structural information required to impart the procognitive activity of the AngIV analog, norleucine(1)-angiotensin IV, resides in its three N-terminal amino acids, Nle-Tyr-Ile. The goal of this project was to chemically modify this tripeptide in such a way to enhance its metabolic stability and barrier permeability to produce a drug candidate with potential clinical utility. Initial results demonstrated that several N- and C-terminal modifications lead to dramatically improved stability while maintaining the capability to reverse scopolamine-induced deficits in Morris water maze performance and augment hippocampal synaptogenesis. Subsequent chemical modifications, which were designed to increase hydrophobicity and decrease hydrogen bonding, yielded an orally active, blood-barrier permeant, metabolically stabilized analog, N-hexanoic-Tyr-Ile-(6) aminohexanoic amide (dihexa), that exhibits excellent antidementia activity in the scopolamine and aged rat models and marked synaptogenic activity. These data suggest that dihexa may have therapeutic potential as a treatment of disorders, such as Alzheimer's disease, where augmented synaptic connectivity may be beneficial.
Collapse
Affiliation(s)
- Alene T McCoy
- Department of Veterinary and Comparative Anatomy, Pharmacology, and Physiology, P.O. Box 6520, Washington State University, Pullman, WA 99164-6520, USA
| | | | | | | | | | | | | | | | | |
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: 15] [Impact Index Per Article: 1.2] [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
|
Kawas LH, Benoist CC, Harding JW, Wayman GA, Abu-Lail NI. Nanoscale mapping of the Met receptor on hippocampal neurons by AFM and confocal microscopy. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2012; 9:428-38. [PMID: 22960190 DOI: 10.1016/j.nano.2012.08.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Revised: 08/10/2012] [Accepted: 08/13/2012] [Indexed: 10/27/2022]
Abstract
UNLABELLED Hepatocyte growth factor (HGF), a neurotrophic protein, acting through its tyrosine kinase receptor, Met, facilitates learning and synaptic plasticity. In concert with the role of the HGF/Met system in synaptic plasticity, we demonstrate that Met is localized to brain regions which undergo extensive synaptic remodeling. We demonstrate that Met activation results in an increase in dendritic spine density and functional synapses. Based on these observations, we hypothesized that Met should be associated with post-synaptic elements found on dendritic spines. Thus, the goal of this study was to determine the sub-cellular localization of Met on hippocampal neurons. Using an atomic force microscopy tip decorated with a specific Met antibody, the location of Met was mapped to different cellular compartments of hippocampal pyramidal neurons. Our results indicated that multimeric activated Met was found to be concentrated in the dendritic compartment while the inactivated monomeric form of Met was prominent on the soma. FROM THE CLINICAL EDITOR The goal of this study was to determine the sub-cellular localization of Met on hippocampal neurons using nanotechnology-based techniques, using an atomic force microscopy tip decorated with a specific Met antibody. The authors demonstrate that multimeric activated Met was found to be concentrated in the dendritic compartment while the inactivated monomeric form of Met was prominent in the soma of hippocampal pyramidal neurons.
Collapse
Affiliation(s)
- Leen H Kawas
- Department of Veterinary and Comparative Anatomy, Pharmacology, and Physiology, Washington State University, Pullman, Washington, USA
| | | | | | | | | |
Collapse
|
28
|
Wright JW, Wilson WL, Wakeling V, Boydstun AS, Jensen A, Kawas L, Harding JW. The Hepatocyte Growth Factor/c-Met Antagonist, Divalinal-Angiotensin IV, Blocks the Acquisition of Methamphetamine Dependent Conditioned Place Preference in Rats. Brain Sci 2012; 2:298-318. [PMID: 24961196 PMCID: PMC4061800 DOI: 10.3390/brainsci2030298] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Revised: 07/06/2012] [Accepted: 08/10/2012] [Indexed: 11/16/2022] Open
Abstract
The use of methamphetamine (MA) is increasing in the U.S. and elsewhere around the world. MA's capacity to cause addiction significantly exceeds other psychostimulant drugs, and its use negatively impacts learning and memory. Recently, attempts have been made to interfere with the presumed mechanism(s) underlying the establishment of drug-induced memory consolidation. The majority of these studies have employed matrix metalloproteinase (MMP) inhibitors to disrupt MMP-induced extracellular matrix molecule dependent synaptic reconfiguration, or GABA receptor agonists. The present investigation utilized an angiotensin IV (AngIV) analogue, Divalinal-AngIV (divalinal), to disrupt acquisition of MA-induced dependence in rats as measured using the conditioned place preference paradigm. Results indicate that both acute and chronic intracerebroventricular infusion of divalinal prior to each daily subcutaneous injection of MA prevented acquisition. However, divalinal was unable to prevent MA-induced reinstatement after prior acquisition followed by extinction trials. These results indicate that prevention of MA dependence can be accomplished by blockade of the brain AT4 receptor subtype. On the other hand, once MA-induced memory consolidation is in place divalinal appears to be ineffective. Mechanistic studies indicated that divalinal is a potent inhibitor of the hepatocyte growth factor (HGF)/c-Met receptor system, and thus it appears that a functional HGF/c-Met system is required for the acquisition of MA-mediated conditioned place preference.
Collapse
Affiliation(s)
- John W Wright
- Department of Psychology, Washington State University, Pullman, WA 99164-4820, USA.
| | - Wendy L Wilson
- Department of Psychology, Dickinson State University, Dickinson, ND 58601, USA.
| | - Vanessa Wakeling
- Department of Psychology, Washington State University, Pullman, WA 99164-4820, USA.
| | - Alan S Boydstun
- L-3 Communications, Link Simulation and Training, Wright Patterson Air Force Base, OH 45433-7955, USA.
| | - Audrey Jensen
- Department of Psychology, Washington State University, Pullman, WA 99164-4820, USA.
| | - Leen Kawas
- Program in Pharmacology and Toxicology, Washington State University, Pullman, WA 99164-6510, USA.
| | - Joseph W Harding
- Department of Psychology, Washington State University, Pullman, WA 99164-4820, USA.
| |
Collapse
|
29
|
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: 160] [Impact Index Per Article: 12.3] [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]
|