1
|
Sharo C, Zhai T, Huang Z. Investigation of Potential Drug Targets Involved in Inflammation Contributing to Alzheimer's Disease Progression. Pharmaceuticals (Basel) 2024; 17:137. [PMID: 38276010 PMCID: PMC10819325 DOI: 10.3390/ph17010137] [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: 04/01/2023] [Revised: 01/15/2024] [Accepted: 01/16/2024] [Indexed: 01/27/2024] Open
Abstract
Alzheimer's disease has become a major public health issue. While extensive research has been conducted in the last few decades, few drugs have been approved by the FDA to treat Alzheimer's disease. There is still an urgent need for understanding the disease pathogenesis, as well as identifying new drug targets for further drug discovery. Alzheimer's disease is known to arise from a build-up of amyloid beta (Aβ) plaques as well as tangles of tau proteins. Along similar lines to Alzheimer's disease, inflammation in the brain is known to stem from the degeneration of tissue and build-up of insoluble materials. A minireview was conducted in this work assessing the genes, proteins, reactions, and pathways that link brain inflammation and Alzheimer's disease. Existing tools in Systems Biology were implemented to build protein interaction networks, mainly for the classical complement pathway and G protein-coupled receptors (GPCRs), to rank the protein targets according to their interactions. The top 10 protein targets were mainly from the classical complement pathway. With the consideration of existing clinical trials and crystal structures, proteins C5AR1 and GARBG1 were identified as the best targets for further drug discovery, through computational approaches like ligand-protein docking techniques.
Collapse
Affiliation(s)
| | | | - Zuyi Huang
- Department of Chemical and Biological Engineering, Villanova University, Villanova, PA 19085, USA
| |
Collapse
|
2
|
Zhou Z, Orchard SG, Nelson MR, Fravel MA, Ernst ME. Angiotensin Receptor Blockers and Cognition: a Scoping Review. Curr Hypertens Rep 2024; 26:1-19. [PMID: 37733162 PMCID: PMC10796582 DOI: 10.1007/s11906-023-01266-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/21/2023] [Indexed: 09/22/2023]
Abstract
PURPOSE OF REVIEW To provide an overview of the association between angiotensin II receptor blocker (ARB) use and cognitive outcomes. RECENT FINDINGS ARBs have previously shown greater neuroprotection compared to other anti-hypertensive classes. The benefits are primarily attributed to the ARB's effect on modulating the renin-angiotensin system via inhibiting the Ang II/AT1R pathway and activating the Ang II/AT2R, Ang IV/AT4R, and Ang-(1-7)/MasR pathways. These interactions are associated with pleiotropic neurocognitive benefits, including reduced β-amyloid accumulation and abnormal hyperphosphorylation of tau, ameliorated brain hypo-fusion, reduced neuroinflammation and synaptic dysfunction, better neurotoxin clearing, and blood-brain barrier function restoration. While ACEis also inhibit AT1R, they simultaneously lower Ang II and block the Ang II/AT2R and Ang IV/AT4R pathways that counterbalance the potential benefits. ARBs may be considered an adjunctive approach for neuroprotection. This preliminary evidence, coupled with their underlying mechanistic pathways, emphasizes the need for future long-term randomized trials to yield more definitive results.
Collapse
Affiliation(s)
- Zhen Zhou
- School of Public Health and Preventive Medicine, Monash University, 553 St Kilda Road, Melbourne, VIC, 3004, Australia.
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia.
| | - Suzanne G Orchard
- School of Public Health and Preventive Medicine, Monash University, 553 St Kilda Road, Melbourne, VIC, 3004, Australia
| | - Mark R Nelson
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Michelle A Fravel
- Department of Pharmacy Practice and Science, College of Pharmacy, The University of Iowa, Iowa, IA, USA
| | - Michael E Ernst
- Department of Pharmacy Practice and Science, College of Pharmacy, The University of Iowa, Iowa, IA, USA.
- Department of Family Medicine, Carver College of Medicine, 01291-A PFP, The University of Iowa, 200 Hawkins Dr, Iowa, IA, 52242, USA.
| |
Collapse
|
3
|
Ali NH, Al‐Kuraishy HM, Al‐Gareeb AI, Alnaaim SA, Alexiou A, Papadakis M, Khalifa AA, Saad HM, Batiha GE. Neprilysin inhibitors and risk of Alzheimer's disease: A future perspective. J Cell Mol Med 2024; 28:e17993. [PMID: 37847125 PMCID: PMC10826440 DOI: 10.1111/jcmm.17993] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 09/15/2023] [Accepted: 10/05/2023] [Indexed: 10/18/2023] Open
Abstract
Alzheimer's disease (AD) is a heterogeneous neurodegenerative disease with multifaceted neuropathological disorders. AD is characterized by intracellular accumulation of phosphorylated tau proteins and extracellular deposition of amyloid beta (Aβ). Various protease enzymes, including neprilysin (NEP), are concerned with the degradation and clearance of Aβ. Indeed, a defective neuronal clearance pathway due to the dysfunction of degradation enzymes might be a possible mechanism for the accumulation of Aβ and subsequent progression of AD neuropathology. NEP is one of the most imperative metalloproteinase enzymes involved in the clearance of Aβ. This review aimed to highlight the possible role of NEP inhibitors in AD. The combination of sacubitril and valsartan which is called angiotensin receptor blocker and NEP inhibitor (ARNI) may produce beneficial and deleterious effects on AD neuropathology. NEP inhibitors might increase the risk of AD by the inhibition of Aβ clearance, and increase brain bradykinin (BK) and natriuretic peptides (NPs), which augment the pathogenesis of AD. These verdicts come from animal model studies, though they may not be applied to humans. However, clinical studies revealed promising safety findings regarding the use of ARNI. Moreover, NEP inhibition increases various neuroprotective peptides involved in inflammation, glucose homeostasis and nerve conduction. Also, NEP inhibitors may inhibit dipeptidyl peptidase 4 (DPP4) expression, ameliorating insulin and glucagon-like peptide 1 (GLP-1) levels. These findings proposed that NEP inhibitors may have a protective effect against AD development by increasing GLP-1, neuropeptide Y (NPY) and substance P, and deleterious effects by increasing brain BK. Preclinical and clinical studies are recommended in this regard.
Collapse
Affiliation(s)
- Naif H. Ali
- Department of Internal Medicine, Medical CollegeNajran UniversityNajranSaudi Arabia
| | - Hayder M. Al‐Kuraishy
- Department of Clinical Pharmacology and Medicine, College of MedicineMustansiriyah UniversityBaghdadIraq
| | - Ali I. Al‐Gareeb
- Department of Clinical Pharmacology and Medicine, College of MedicineMustansiriyah UniversityBaghdadIraq
| | - Saud A. Alnaaim
- Clinical Neurosciences Department, College of MedicineKing Faisal UniversityHofufSaudi Arabia
| | - Athanasios Alexiou
- Department of Science and EngineeringNovel Global Community Educational FoundationHebershamNew South WalesAustralia
- AFNP MedWienAustria
| | - Marios Papadakis
- Department of Surgery IIUniversity Hospital Witten‐Herdecke, University of Witten‐HerdeckeWuppertalGermany
| | - Asmaa A. Khalifa
- Department of Pharmacology and Therapeutics, Faculty of PharmacyPharos University in AlexandriaAlexandriaEgypt
| | - Hebatallah M. Saad
- Department of Pathology, Faculty of Veterinary MedicineMatrouh UniversityMatrouhEgypt
| | - Gaber El‐Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary MedicineDamanhour UniversityDamanhourAlBeheiraEgypt
| |
Collapse
|
4
|
Identification of Human Kinin-Forming Enzyme Inhibitors from Medicinal Herbs. Molecules 2021; 26:molecules26144126. [PMID: 34299400 PMCID: PMC8307503 DOI: 10.3390/molecules26144126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 06/26/2021] [Accepted: 07/02/2021] [Indexed: 11/18/2022] Open
Abstract
The goal of this study was to assess the pharmacological effects of black tea (Camellia sinensis var. assamica) water extract on human kinin-forming enzymes in vitro. Tea is a highly consumed beverage in the world. Factor XII (FXII, Hageman factor)-independent- and -dependent activation of prekallikrein to kallikrein leads to the liberation of bradykinin (BK) from high-molecular-weight kininogen (HK). The excessive BK production causes vascular endothelial and nonvascular smooth muscle cell permeability, leading to angioedema. The prevalence of angiotensin-converting enzyme inhibitor (ACEI)-induced angioedema appears to be through BK. Both histamine and BK are potent inflammatory mediators. However, the treatments for histamine-mediated angioedema are unsuitable for BK-mediated angioedema. We hypothesized that long-term consumption of tea would reduce bradykinin-dependent processes within the systemic and pulmonary vasculature, independent of the anti-inflammatory actions of polyphenols. A purified fraction of the black tea water extract inhibited both kallikrein and activated FXII. The black tea water extracts inhibited factor XII-induced cell migration and inhibited the production of kallikrein on the endothelial cell line. We compared the inhibitory effects of the black tea water extract and twenty-three well-known anti-inflammatory medicinal herbs, in inhibiting both kallikrein and FXII. Surprisingly, arjunglucoside II specifically inhibited the activated factor XII (FXIIa), but not the kallikrein and the activated factor XI. Taken together, the black tea water extract exerts its anti-inflammatory effects, in part, by inhibiting kallikrein and activated FXII, which are part of the plasma kallikrein–kinin system (KKS), and by decreasing BK production. The inhibition of kallikrein and activated FXII represents a unique polyphenol-independent anti-inflammatory mechanism of action for the black tea.
Collapse
|
5
|
de Souza Maciel I, Azevedo VM, Oliboni P, Campos MM. Blockade of the kinin B 1 receptor counteracts the depressive-like behaviour and mechanical allodynia in ovariectomised mice. Behav Brain Res 2021; 412:113439. [PMID: 34197868 DOI: 10.1016/j.bbr.2021.113439] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 05/31/2021] [Accepted: 06/25/2021] [Indexed: 12/27/2022]
Abstract
Menopause is related to a decline in ovarian oestrogen production, affecting the perception of the somatosensory stimuli, changing the immune-inflammatory systems, and triggering depressive symptoms. It has been demonstrated that the inhibition of the kinin B1 and B2 receptors (B1R and B2R) prevented the depressive-like behaviour and the mechanical allodynia that was induced by immune-inflammatory mediators in mice. However, there is no evidence regarding the role of the kinin receptors in the depressive-like and nociceptive behaviour in female mice that were subjected to bilateral ovariectomy (OVX). This study has shown that the OVX mice developed time-related mechanical allodynia, together with an increased immobility time as indicative of depression. Both of these changes were reduced by the genetic deletion of B1R, or by the pharmacological blockade of the selective kinin B1R antagonist R-715 (acute, i.p.). The genetic deletion or the pharmacological inhibition of B2R (HOE 140, i.p.) did not prevent the OVX-elicited behavioural changes. The data has suggested a particular modulation of kinin B1R in the nociceptive and depressive-like behaviour in the OVX mice. The selective inhibition of the B1R receptor may be a new pharmacological target for treating pain and depression symptoms in women during the perimenopause/menopause period.
Collapse
Affiliation(s)
- Izaque de Souza Maciel
- Programa de Pós-graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil; Centro de Pesquisa em Toxicologia e Farmacologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil; Escola de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil.
| | - Vanessa Machado Azevedo
- Centro de Pesquisa em Toxicologia e Farmacologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Patricia Oliboni
- Centro de Pesquisa em Toxicologia e Farmacologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil; Programa de Pós-graduação em Odontologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Maria Martha Campos
- Programa de Pós-graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil; Centro de Pesquisa em Toxicologia e Farmacologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil; Programa de Pós-graduação em Odontologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
6
|
Petrella C, Ciotti MT, Nisticò R, Piccinin S, Calissano P, Capsoni S, Mercanti D, Cavallaro S, Possenti R, Severini C. Involvement of Bradykinin Receptor 2 in Nerve Growth Factor Neuroprotective Activity. Cells 2020; 9:cells9122651. [PMID: 33321704 PMCID: PMC7763563 DOI: 10.3390/cells9122651] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/03/2020] [Accepted: 12/07/2020] [Indexed: 11/17/2022] Open
Abstract
Neurotrophin nerve growth factor (NGF) has been demonstrated to upregulate the gene expression of bradykinin receptor 2 (B2R) on sensory neurons, thus facilitating nociceptive signals. The aim of the present study is to investigate the involvement of B2R in the NGF mechanism of action in nonsensory neurons in vitro by using rat mixed cortical primary cultures (CNs) and mouse hippocampal slices, and in vivo in Alzheimer’s disease (AD) transgenic mice (5xFAD) chronically treated with NGF. A significant NGF-mediated upregulation of B2R was demonstrated by microarray, Western blot, and immunofluorescence analysis in CNs, indicating microglial cells as the target of this modulation. The B2R involvement in the NGF mechanism of action was also demonstrated by using a selective B2R antagonist which was able to reverse the neuroprotective effect of NGF in CNs, as revealed by viability assay, and the NGF-induced long-term potentiation (LTP) in hippocampal slices. To confirm in vitro observations, B2R upregulation was observed in 5xFAD mouse brain following chronic intranasal NGF treatment. This study demonstrates for the first time that B2R is a key element in the neuroprotective activity and synaptic plasticity mediated by NGF in brain cells.
Collapse
Affiliation(s)
- Carla Petrella
- Institute of Biochemistry and Cell Biology, National Research Council, Sapienza University of Rome, Viale del Policlinico, 155-00161 Rome, Italy; (C.P.); (M.T.C.); (D.M.)
| | - Maria Teresa Ciotti
- Institute of Biochemistry and Cell Biology, National Research Council, Sapienza University of Rome, Viale del Policlinico, 155-00161 Rome, Italy; (C.P.); (M.T.C.); (D.M.)
| | - Robert Nisticò
- Department of Biology, University of Rome “Tor Vergata”, Via della Ricerca Scientifica 1, 00133 Rome, Italy; (R.N.); (S.P.)
- Rita Levi-Montalcini European Brain Research Institute (EBRI), Viale Regina Elena, 295, 00161 Rome, Italy;
| | - Sonia Piccinin
- Department of Biology, University of Rome “Tor Vergata”, Via della Ricerca Scientifica 1, 00133 Rome, Italy; (R.N.); (S.P.)
| | - Pietro Calissano
- Rita Levi-Montalcini European Brain Research Institute (EBRI), Viale Regina Elena, 295, 00161 Rome, Italy;
| | - Simona Capsoni
- Section of Physiology, Department of Biomedical and Specialty Surgical Sciences, University of Ferrara, Via Luigi Borsari 46, 44121 Ferrara, Italy;
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, Piazza dei Cavalieri, 7, 56126 Pisa, Italy
| | - Delio Mercanti
- Institute of Biochemistry and Cell Biology, National Research Council, Sapienza University of Rome, Viale del Policlinico, 155-00161 Rome, Italy; (C.P.); (M.T.C.); (D.M.)
| | - Sebastiano Cavallaro
- Institute for Biomedical Research and Innovation, National Research Council, Via Paolo Gaifami 18, 95126 Catania, Italy;
| | - Roberta Possenti
- Department Medicine of Systems, University of Rome “Tor Vergata”, Via della Ricerca Scientifica 1, 00133 Rome, Italy;
| | - Cinzia Severini
- Institute of Biochemistry and Cell Biology, National Research Council, Sapienza University of Rome, Viale del Policlinico, 155-00161 Rome, Italy; (C.P.); (M.T.C.); (D.M.)
- Correspondence:
| |
Collapse
|
7
|
Atorvastatin Improves Mitochondrial Function and Prevents Oxidative Stress in Hippocampus Following Amyloid-β 1-40 Intracerebroventricular Administration in Mice. Mol Neurobiol 2020; 57:4187-4201. [PMID: 32683653 DOI: 10.1007/s12035-020-02026-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 07/14/2020] [Indexed: 12/12/2022]
Abstract
Amyloid-β (Aβ) peptides play a significant role in the pathogenesis of Alzheimer's disease (AD). Neurotoxic effects promoted by Aβ peptides involve glutamate transmission impairment, decrease of neurotrophic factors, mitochondrial dysfunction, oxidative stress, synaptotoxicity, and neuronal degeneration. Here, we assessed the early events evoked by Aβ1-40 on the hippocampus. Additionally, we sought to unravel the molecular mechanisms of atorvastatin preventive effect on Aβ-induced hippocampal damage. Mice were treated orally (p.o.) with atorvastatin 10 mg/kg/day during 7 consecutive days before the intracerebroventricular (i.c.v.) infusion of Aβ1-40 (400 pmol/site). Twenty-four hours after Aβ1-40 infusion, a reduced content of mature BDNF/proBDNF ratio was observed in Aβ-treated mice. However, there is no alteration in synaptophysin, PSD-95, and doublecortin immunocontent in the hippocampus. Aβ1-40 promoted an increase in reactive oxygen species (ROS) and nitric oxide (NO) generation in hippocampal slices, and atorvastatin prevented this oxidative burst. Mitochondrial OXPHOS was measured by high-resolution respirometry. At this time point, Aβ1-40 did not alter the O2 consumption rates (OCR) related to phosphorylating state associated with complexes I and II, and the maximal OCR. However, atorvastatin increased OCR of phosphorylating state associated with complex I and complexes I and II, maximal OCR of complexes I and II, and OCR associated with mitochondrial spare capacity. Atorvastatin treatment improved mitochondrial function in the rodent hippocampus, even after Aβ infusion, pointing to a promising effect of improving brain mitochondria bioenergetics. Therefore, atorvastatin could act as an adjuvant in battling the symptoms of AD to preventing or delaying the disease progression.
Collapse
|
8
|
Singh PK, Chen ZL, Ghosh D, Strickland S, Norris EH. Increased plasma bradykinin level is associated with cognitive impairment in Alzheimer's patients. Neurobiol Dis 2020; 139:104833. [PMID: 32173555 PMCID: PMC7175647 DOI: 10.1016/j.nbd.2020.104833] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 02/28/2020] [Accepted: 03/11/2020] [Indexed: 12/16/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by the presence of proteinaceous brain deposits, brain atrophy, vascular dysfunction, and chronic inflammation. Along with cerebral inflammation, peripheral inflammation is also evident in many AD patients. Bradykinin, a proinflammatory plasma peptide, is also linked to AD pathology. For example, bradykinin infusion into the hippocampus causes learning and memory deficits in rats, and blockade of the bradykinin receptor lessens cognitive impairment in AD mouse models. Even though it has been hypothesized that plasma bradykinin could contribute to inflammation in AD, the level of plasma bradykinin and its association with beta-amyloid (Aβ) pathology in AD patients had not been explored. Here, we assessed plasma bradykinin levels in AD patients and age-matched non-demented (ND) control individuals. We found significantly elevated plasma bradykinin levels in AD patients compared to ND subjects. Additionally, changes in plasma bradykinin levels were more profound in many AD patients with severe cognitive impairment, suggesting that peripheral bradykinin could play a role in dementia most likely via inflammation. Bradykinin levels in the cerebrospinal fluid (CSF) were reduced in AD patients and exhibited an inverse correlation with the CSF Aβ40/Aβ42 ratio. We also report that bradykinin interacts with the fibrillar form of Aβ and co-localizes with Aβ plaques in the post-mortem human AD brain. These findings connect the peripheral inflammatory pathway to cerebral abnormalities and identify a novel mechanism of inflammatory pathology in AD.
Collapse
Affiliation(s)
- Pradeep K Singh
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY 10065, USA
| | - Zu-Lin Chen
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY 10065, USA
| | - Dhiman Ghosh
- Laboratory of Physical Chemistry, ETH Zürich, WolfgangPauli-Str. 10, 8093 Zürich, Switzerland
| | - Sidney Strickland
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY 10065, USA
| | - Erin H Norris
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
9
|
Ji B, Wang Q, Xue Q, Li W, Li X, Wu Y. The Dual Role of Kinin/Kinin Receptors System in Alzheimer's Disease. Front Mol Neurosci 2019; 12:234. [PMID: 31632239 PMCID: PMC6779775 DOI: 10.3389/fnmol.2019.00234] [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: 05/31/2019] [Accepted: 09/13/2019] [Indexed: 11/30/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common neurodegenerative disease characterized by progressive spatial disorientation, learning and memory deficits, responsible for 60%–80% of all dementias. However, the pathological mechanism of AD remains unknown. Numerous studies revealed that kinin/kinin receptors system (KKS) may be involved in the pathophysiology of AD. In this review article, we summarized the roles of KKS in neuroinflammation, cerebrovascular impairment, tau phosphorylation, and amyloid β (Aβ) generation in AD. Moreover, we provide new insights into the mechanistic link between KKS and AD, and highlight the KKS as a potential therapeutic target for AD treatment.
Collapse
Affiliation(s)
- Bingyuan Ji
- Neurobiology Institute, School of Mental Health, Jining Medical University, Jining, China
| | - Qinqin Wang
- Neurobiology Institute, School of Mental Health, Jining Medical University, Jining, China
| | - Qingjie Xue
- Department of Pathogenic Biology, Jining Medical University, Jining, China
| | - Wenfu Li
- Neurobiology Institute, School of Mental Health, Jining Medical University, Jining, China
| | - Xuezhi Li
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, China.,Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, China
| | - Yili Wu
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, China.,Shandong Key Laboratory of Behavioral Medicine, School of Mental Health, Jining Medical University, Jining, China
| |
Collapse
|
10
|
Petrella C, Di Certo MG, Barbato C, Gabanella F, Ralli M, Greco A, Possenti R, Severini C. Neuropeptides in Alzheimer’s Disease: An Update. Curr Alzheimer Res 2019; 16:544-558. [DOI: 10.2174/1567205016666190503152555] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 03/19/2019] [Accepted: 04/30/2019] [Indexed: 12/19/2022]
Abstract
Neuropeptides are small proteins broadly expressed throughout the central nervous system, which act as neurotransmitters, neuromodulators and neuroregulators. Growing evidence has demonstrated the involvement of many neuropeptides in both neurophysiological functions and neuropathological conditions, among which is Alzheimer’s disease (AD). The role exerted by neuropeptides in AD is endorsed by the evidence that they are mainly neuroprotective and widely distributed in brain areas responsible for learning and memory processes. Confirming this point, it has been demonstrated that numerous neuropeptide-containing neurons are pathologically altered in brain areas of both AD patients and AD animal models. Furthermore, the levels of various neuropeptides have been found altered in both Cerebrospinal Fluid (CSF) and blood of AD patients, getting insights into their potential role in the pathophysiology of AD and offering the possibility to identify novel additional biomarkers for this pathology. We summarized the available information about brain distribution, neuroprotective and cognitive functions of some neuropeptides involved in AD. The main focus of the current review was directed towards the description of clinical data reporting alterations in neuropeptides content in both AD patients and AD pre-clinical animal models. In particular, we explored the involvement in the AD of Thyrotropin-Releasing Hormone (TRH), Cocaine- and Amphetamine-Regulated Transcript (CART), Cholecystokinin (CCK), bradykinin and chromogranin/secretogranin family, discussing their potential role as a biomarker or therapeutic target, leaving the dissertation of other neuropeptides to previous reviews.
Collapse
Affiliation(s)
- Carla Petrella
- Department of Sense Organs, CNR, Institute of Cell Biology and Neurobiology, University Sapienza of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Maria Grazia Di Certo
- Department of Sense Organs, CNR, Institute of Cell Biology and Neurobiology, University Sapienza of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Christian Barbato
- Department of Sense Organs, CNR, Institute of Cell Biology and Neurobiology, University Sapienza of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Francesca Gabanella
- Department of Sense Organs, CNR, Institute of Cell Biology and Neurobiology, University Sapienza of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Massimo Ralli
- Department of Sense Organs, University Sapienza of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Antonio Greco
- Department of Sense Organs, University Sapienza of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Roberta Possenti
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Cinzia Severini
- Department of Sense Organs, CNR, Institute of Cell Biology and Neurobiology, University Sapienza of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| |
Collapse
|
11
|
Adalimumab improves cognitive impairment, exerts neuroprotective effects and attenuates neuroinflammation in an Aβ1-40-injected mouse model of Alzheimer's disease. Cytotherapy 2019; 21:671-682. [DOI: 10.1016/j.jcyt.2019.04.054] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 04/11/2019] [Accepted: 04/11/2019] [Indexed: 01/11/2023]
|
12
|
Ni R, Kindler DR, Waag R, Rouault M, Ravikumar P, Nitsch R, Rudin M, Camici GG, Liberale L, Kulic L, Klohs J. fMRI Reveals Mitigation of Cerebrovascular Dysfunction by Bradykinin Receptors 1 and 2 Inhibitor Noscapine in a Mouse Model of Cerebral Amyloidosis. Front Aging Neurosci 2019; 11:27. [PMID: 30890928 PMCID: PMC6413713 DOI: 10.3389/fnagi.2019.00027] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 01/30/2019] [Indexed: 11/28/2022] Open
Abstract
Functional magnetic resonance imaging (fMRI) techniques can be used to assess cerebrovascular dysfunction in Alzheimer’s disease, an important and early contributor to pathology. We hypothesized that bradykinin receptor inhibition alleviates the vascular dysfunction in a transgenic arcAβ mouse model of cerebral amyloidosis and that fMRI techniques can be used to monitor the treatment response. Transgenic arcAβ mice, and non-transgenic littermates of 14 months-of-age were either treated with the bradykinin receptors 1 and 2 blocker noscapine or received normal drinking water as control over 3 months (n = 8–11/group) and all mice were assessed using fMRI at the end of the treatment period. Perfusion MRI using an arterial spin labeling technique showed regional hypoperfusion in arcAβ compared to non-transgenic controls, which was alleviated by noscapine treatment. Similarly, measuring cerebral blood volume changes upon pharmacological stimulation using vessel dilator acetazolamide revealed recovery of regional impairment of cerebral vascular reactivity in arcAβ mice upon noscapine treatment. In addition, we assessed with immunohistochemistry beta-amyloid (Aβ) and inflammation levels in brain sections. Immunohistological stainings for Aβ deposition (6E10) and related microgliosis (Iba1) in the cortex and hippocampus were found comparable between noscapine-treated and untreated arcAβ mice. In addition, levels of soluble and insoluble Aβ38, Aβ40, Aβ42 were found to be similar in brain tissue homogenates of noscapine-treated and untreated arcAβ mice using electro-chemiluminescent based immunoassay. In summary, bradykinin receptors blockade recovered cerebral vascular dysfunction in a mouse model of cerebral amyloidosis. fMRI methods revealed the functional deficit in disease condition and were useful tools to monitor the treatment response.
Collapse
Affiliation(s)
- Ruiqing Ni
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zürich, Switzerland.,Zurich Neuroscience Center, Zürich, Switzerland
| | - Diana Rita Kindler
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zürich, Switzerland
| | - Rebecca Waag
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zürich, Switzerland
| | - Marie Rouault
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zürich, Switzerland.,Zurich Neuroscience Center, Zürich, Switzerland
| | - Priyanka Ravikumar
- Institute for Regenerative Medicine, University of Zurich, Zürich, Switzerland
| | - Roger Nitsch
- Institute for Regenerative Medicine, University of Zurich, Zürich, Switzerland
| | - Markus Rudin
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zürich, Switzerland
| | - Giovanni G Camici
- Zurich Neuroscience Center, Zürich, Switzerland.,Center for Molecular Cardiology, University of Zurich, Zürich, Switzerland
| | - Luca Liberale
- Institute for Regenerative Medicine, University of Zurich, Zürich, Switzerland.,Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Luka Kulic
- Zurich Neuroscience Center, Zürich, Switzerland.,Institute for Regenerative Medicine, University of Zurich, Zürich, Switzerland
| | - Jan Klohs
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zürich, Switzerland.,Zurich Neuroscience Center, Zürich, Switzerland
| |
Collapse
|
13
|
Medeiros ADM, Silva RH. Sex Differences in Alzheimer’s Disease: Where Do We Stand? J Alzheimers Dis 2019; 67:35-60. [DOI: 10.3233/jad-180213] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- André de Macêdo Medeiros
- Behavioral Neuroscience Laboratory, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
- Center of Health and Biological Sciences, Universidade Federal Rural do Semiárido, Mossoró, Brazil
| | - Regina Helena Silva
- Behavioral Neuroscience Laboratory, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
14
|
Petek B, Villa-Lopez M, Loera-Valencia R, Gerenu G, Winblad B, Kramberger MG, Ismail MAM, Eriksdotter M, Garcia-Ptacek S. Connecting the brain cholesterol and renin-angiotensin systems: potential role of statins and RAS-modifying medications in dementia. J Intern Med 2018; 284:620-642. [PMID: 30264910 DOI: 10.1111/joim.12838] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Millions of people worldwide receive agents targeting the renin-angiotensin system (RAS) to treat hypertension or statins to lower cholesterol. The RAS and cholesterol metabolic pathways in the brain are autonomous from their systemic counterparts and are interrelated through the cholesterol metabolite 27-hydroxycholesterol (27-OHC). These systems contribute to memory and dementia pathogenesis through interference in the amyloid-beta cascade, vascular mechanisms, glucose metabolism, apoptosis, neuroinflammation and oxidative stress. Previous studies examining the relationship between these treatments and cognition and dementia risk have produced inconsistent results. Defining the blood-brain barrier penetration of these medications has been challenging, and the mechanisms of action on cognition are not clearly established. Potential biases are apparent in epidemiological and clinical studies, such as reverse epidemiology, indication bias, problems defining medication exposure, uncertain and changing doses, and inappropriate grouping of outcomes and medications. This review summarizes current knowledge of the brain cholesterol and RAS metabolism and the mechanisms by which these pathways affect neurodegeneration. The putative mechanisms of action of statins and medications inhibiting the RAS will be examined, together with prior clinical and animal studies on their effects on cognition. We review prior epidemiological studies, analysing their strengths and biases, and identify areas for future research. Understanding the pathophysiology of the brain cholesterol system and RAS and their links to neurodegeneration has enormous potential. In future, well-designed epidemiological studies could identify potential treatments for Alzheimer's disease (AD) amongst medications that are already in use for other indications.
Collapse
Affiliation(s)
- B Petek
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.,Department of Neurology, University Medical Centre, Ljubljana, Slovenia.,University of Ljubljana, Ljubljana, Slovenia
| | - M Villa-Lopez
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - R Loera-Valencia
- Center for Alzheimer Research, Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - G Gerenu
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.,Department of Neurosciences, Biodonostia Health Research Institute, San Sebastian, Spain.,Center for Networked Biomedical Research in Neurodegenerative Diseases, CIBERNED, Health Institute Carlos III, Ministry of Economy and Competitiveness, Madrid, Spain
| | - B Winblad
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.,Department of Geriatric Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - M G Kramberger
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.,Department of Neurology, University Medical Centre, Ljubljana, Slovenia.,University of Ljubljana, Ljubljana, Slovenia
| | - M-A-M Ismail
- Center for Alzheimer Research, Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.,Theme Neuro, Diseases of the Nervous System patient flow, Karolinska University Hospital, Huddinge, Sweden
| | - M Eriksdotter
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.,Department of Geriatric Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - S Garcia-Ptacek
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.,Department of Internal Medicine, Neurology Section, Södersjukhuset, Stockholm, Sweden
| |
Collapse
|
15
|
Bradykinin B2 receptor is essential to running-induced cell proliferation in the adult mouse hippocampus. Brain Struct Funct 2018; 223:3901-3907. [DOI: 10.1007/s00429-018-1711-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 07/04/2018] [Indexed: 12/21/2022]
|
16
|
Nokkari A, Abou-El-Hassan H, Mechref Y, Mondello S, Kindy MS, Jaffa AA, Kobeissy F. Implication of the Kallikrein-Kinin system in neurological disorders: Quest for potential biomarkers and mechanisms. Prog Neurobiol 2018; 165-167:26-50. [PMID: 29355711 PMCID: PMC6026079 DOI: 10.1016/j.pneurobio.2018.01.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 01/15/2018] [Indexed: 01/06/2023]
Abstract
Neurological disorders represent major health concerns in terms of comorbidity and mortality worldwide. Despite a tremendous increase in our understanding of the pathophysiological processes involved in disease progression and prevention, the accumulated knowledge so far resulted in relatively moderate translational benefits in terms of therapeutic interventions and enhanced clinical outcomes. Aiming at specific neural molecular pathways, different strategies have been geared to target the development and progression of such disorders. The kallikrein-kinin system (KKS) is among the most delineated candidate systems due to its ubiquitous roles mediating several of the pathophysiological features of these neurological disorders as well as being implicated in regulating various brain functions. Several experimental KKS models revealed that the inhibition or stimulation of the two receptors of the KKS system (B1R and B2R) can exhibit neuroprotective and/or adverse pathological outcomes. This updated review provides background details of the KKS components and their functions in different neurological disorders including temporal lobe epilepsy, traumatic brain injury, stroke, spinal cord injury, Alzheimer's disease, multiple sclerosis and glioma. Finally, this work will highlight the putative roles of the KKS components as potential neurotherapeutic targets and provide future perspectives on the possibility of translating these findings into potential clinical biomarkers in neurological disease.
Collapse
Affiliation(s)
- Amaly Nokkari
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Lebanon
| | - Hadi Abou-El-Hassan
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Yehia Mechref
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX, USA
| | - Stefania Mondello
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Mark S Kindy
- Department of Pharmaceutical Science, College of Pharmacy, University of South Florida, Tampa, FL, USA; James A. Haley VA Medical Center, Tampa, FL, USA
| | - Ayad A Jaffa
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Lebanon; Department of Medicine, Medical University of South, Charleston, SC, USA.
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Lebanon; Center for Neuroproteomics & Biomarkers Research, Department of Psychiatry, McKnight Brain Institute, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
17
|
Zang X, Cheng ZY, Sun Y, Hua N, Zhu LH, He L. The ameliorative effects and underlying mechanisms of dopamine D1-like receptor agonist SKF38393 on Aβ 1-42-induced cognitive impairment. Prog Neuropsychopharmacol Biol Psychiatry 2018; 81:250-261. [PMID: 28939187 DOI: 10.1016/j.pnpbp.2017.09.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 08/30/2017] [Accepted: 09/18/2017] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disease characterized by extracellular amyloid plaques and intracellular neurofibrillary tangles. It is the most common form of human cognitive decline and dementia. In this study, we aim to systematically investigate the ameliorative effects of dopamine D1-like receptor agonist SKF38393 on cognitive dysfunction and explore its underlying mechanisms. The Aβ1-42 was injected intracerebroventricularly to establish cognitive disorder model. Then, a series of behavior tests were used. In order to further study the mechanisms, some relevant protein was assessed by ELISA method and Western blot. The results in behavior tests revealed that SKF38393 significantly ameliorated all the test indexes compared with the model mice. Then SKF38393 increased phosphorylation of cAMP response element binding protein (CREB) and expression of Bcl-2 in Western blot analyses. Furthermore, in ELISA assay, SKF38393 significantly increased the brain-derived neurotrophic factor (BDNF) levels and reduced the β-site APP cleaving enzyme1 (BACE1) and Aβ1-42 levels in hippocampus and cortex of mice. However, compared with SKF38393-H, all these results were significantly reversed by the dopamine D1 receptor antagonist SCH23390. These results indicated that SKF38393 could ameliorate Aβ1-42-induced cognitive dysfunction in mice, which may be related to D1 receptor activation. It leads to the phosphorylation of CREB, which promote the expression of BDNF, Bcl-2 and decrease the expression of Aβ1-42 of mice. Our findings suggest that dopamine D1-like receptor may be a potential target for the treatment of AD and its agonists may become a novel drug in the future.
Collapse
Affiliation(s)
- Xuan Zang
- Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China
| | - Zhao-Yan Cheng
- Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China
| | - Yi Sun
- Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China
| | - Nan Hua
- Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China
| | - Li-Hua Zhu
- Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China
| | - Ling He
- Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
18
|
Abstract
INTRODUCTION Kinins are peptide mediators exerting their pro-inflammatory actions by the selective stimulation of two distinct G-protein coupled receptors, termed BKB1R and BKB2R. While BKB2R is constitutively expressed in a multitude of tissues, BKB1R is hardly expressed at baseline but highly inducible by inflammatory mediators. In particular, BKB1R was shown to be involved in the pathogenesis of numerous inflammatory diseases. Areas covered: This review intends to evaluate the therapeutic potential of substances interacting with the BKB1R. To this purpose we summarize the published literature on animal studies with antagonists and knockout mice for this receptor. Expert Opinion: In most cases the pharmacological inhibition of BKB1R or its genetic deletion was beneficial for the outcome of the disease in animal models. Therefore, several companies have developed BKB1R antagonists and tested them in phase I and II clinical trials. However, none of the developed BKB1R antagonists was further developed for clinical use. We discuss possible reasons for this failure of translation of preclinical findings on BKB1R antagonists into the clinic.
Collapse
Affiliation(s)
- Fatimunnisa Qadri
- a Max-Delbrück Center for Molecular Medicine (MDC) , Berlin , Germany
| | - Michael Bader
- a Max-Delbrück Center for Molecular Medicine (MDC) , Berlin , Germany.,b Berlin Institute of Health (BIH) , Berlin , Germany.,c Charité University Medicine Berlin , Germany.,d German Center for Cardiovascular Research (DZHK) site Berlin , Berlin , Germany.,e Institute for Biology , University of Lübeck , Lübeck , Germany
| |
Collapse
|
19
|
Naletova I, Nicoletti VG, Milardi D, Pietropaolo A, Grasso G. Copper, differently from zinc, affects the conformation, oligomerization state and activity of bradykinin. Metallomics 2017; 8:750-61. [PMID: 27328010 DOI: 10.1039/c6mt00067c] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The sole role of bradykinin (BK) as an inflammatory mediator is controversial, as recent data also support an anti-inflammatory role for BK in Alzheimer's disease (AD). The involvement of two different receptors (B1R and B2R) could be a key to understand this issue. However, although copper and zinc dyshomeostasis has been demonstrated to be largely involved in the development of AD, a detailed study of the interaction of BK with these two metal ions has never been addressed. In this work, we have applied mass spectrometry, circular dichroism as well as computational methods in order to assess if copper and zinc have the ability to modulate the conformation and oligomerization of BK. In addition, we have correlated the chemical data with the effect of metals on the activity of BK analyzed in cell cultures by biochemical procedures. The biochemical analyses on monocyte/macrophage cell culture (THP-1 Cell Line human) in line with the effect of metals on the conformation of BK showed that the presence of copper can affect the signaling cascade mediated by the BK receptors. The results obtained show a further role of metal ions, particularly copper, in the development and outcome of neuroinflammatory diseases. The possible implications in AD are discussed.
Collapse
Affiliation(s)
- Irina Naletova
- Dipartimento di Scienze Biomediche e Biotecnologiche "BIOMETEC", Università degli Studi di Catania, Via S. Sofia 64, 95125 Catania, Italy. and Consorzio Interuniversitario di Ricerca in Chimica dei Metalli nei Sistemi Biologici (C.I.R.C.M.S.B.), Piazza Umberto I, 1-70121 Bari, Italy
| | - Vincenzo G Nicoletti
- Dipartimento di Scienze Biomediche e Biotecnologiche "BIOMETEC", Università degli Studi di Catania, Via S. Sofia 64, 95125 Catania, Italy. and Consorzio Interuniversitario di Ricerca in Chimica dei Metalli nei Sistemi Biologici (C.I.R.C.M.S.B.), Piazza Umberto I, 1-70121 Bari, Italy
| | - Danilo Milardi
- Istituto di Biostrutture e Bioimmagini, Consiglio Nazionale delle Ricerche, Via Paolo Gaifami 18, 95126 Catania, Italy
| | - Adriana Pietropaolo
- Dipartimento di Scienze della Salute, Università di Catanzaro, Campus Universitario, Viale Europa, 88100 Catanzaro, Italy
| | - Giuseppe Grasso
- Dipartimento di Scienze Chimiche, Università degli Studi di Catania, Viale Andrea Doria 6, 95125, Catania, Italy.
| |
Collapse
|
20
|
Simino J, Wang Z, Bressler J, Chouraki V, Yang Q, Younkin SG, Seshadri S, Fornage M, Boerwinkle E, Mosley TH. Whole exome sequence-based association analyses of plasma amyloid-β in African and European Americans; the Atherosclerosis Risk in Communities-Neurocognitive Study. PLoS One 2017; 12:e0180046. [PMID: 28704393 PMCID: PMC5509141 DOI: 10.1371/journal.pone.0180046] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 06/08/2017] [Indexed: 12/30/2022] Open
Abstract
Objective We performed single-variant and gene-based association analyses of plasma amyloid-β (aβ) concentrations using whole exome sequence from 1,414 African and European Americans. Our goal was to identify genes that influence plasma aβ42 concentrations and aβ42:aβ40 ratios in late middle age (mean = 59 years), old age (mean = 77 years), or change over time (mean = 18 years). Methods Plasma aβ measures were linearly regressed onto age, gender, APOE ε4 carrier status, and time elapsed between visits (fold-changes only) separately by race. Following inverse normal transformation of the residuals, seqMeta was used to conduct race-specific single-variant and gene-based association tests while adjusting for population structure. Linear regression models were fit on autosomal variants with minor allele frequencies (MAF)≥1%. T5 burden and Sequence Kernel Association (SKAT) gene-based tests assessed functional variants with MAF≤5%. Cross-race fixed effects meta-analyses were Bonferroni-corrected for the number of variants or genes tested. Results Seven genes were associated with aβ in late middle age or change over time; no associations were identified in old age. Single variants in KLKB1 (rs3733402; p = 4.33x10-10) and F12 (rs1801020; p = 3.89x10-8) were significantly associated with midlife aβ42 levels through cross-race meta-analysis; the KLKB1 variant replicated internally using 1,014 additional participants with exome chip. ITPRIP, PLIN2, and TSPAN18 were associated with the midlife aβ42:aβ40 ratio via the T5 test; TSPAN18 was significant via the cross-race meta-analysis, whereas ITPRIP and PLIN2 were European American-specific. NCOA1 and NT5C3B were associated with the midlife aβ42:aβ40 ratio and the fold-change in aβ42, respectively, via SKAT in African Americans. No associations replicated externally (N = 725). Conclusion We discovered age-dependent genetic effects, established associations between vascular-related genes (KLKB1, F12, PLIN2) and midlife plasma aβ levels, and identified a plausible Alzheimer’s Disease candidate gene (ITPRIP) influencing cell death. Plasma aβ concentrations may have dynamic biological determinants across the lifespan; plasma aβ study designs or analyses must consider age.
Collapse
Affiliation(s)
- Jeannette Simino
- Gertrude C. Ford MIND Center, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
- Department of Data Science, John D. Bower School of Population Health, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
- * E-mail:
| | - Zhiying Wang
- Human Genetics Center, Department of Epidemiology, Human Genetics & Environmental Sciences, School of Public Health, University of Texas Health Science Center, Houston, Texas, United States of America
| | - Jan Bressler
- Human Genetics Center, Department of Epidemiology, Human Genetics & Environmental Sciences, School of Public Health, University of Texas Health Science Center, Houston, Texas, United States of America
| | - Vincent Chouraki
- Lille University, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk factors and molecular determinants of aging-related diseases; Lille, France
| | - Qiong Yang
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, United States of America
- The National Heart, Lung, and Blood Institute’s Framingham Heart Study, Framingham, Massachusetts, United States of America
| | - Steven G. Younkin
- Department of Neuroscience, Mayo Clinic College of Medicine, Mayo Clinic Jacksonville, Jacksonville, Florida, United States of America
| | - Sudha Seshadri
- The National Heart, Lung, and Blood Institute’s Framingham Heart Study, Framingham, Massachusetts, United States of America
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Myriam Fornage
- Human Genetics Center, Department of Epidemiology, Human Genetics & Environmental Sciences, School of Public Health, University of Texas Health Science Center, Houston, Texas, United States of America
- The Brown Foundation Institute of Molecular Medicine, Research Center for Human Genetics, The University of Texas Health Science Center, Houston, Texas, United States of America
| | - Eric Boerwinkle
- Human Genetics Center, Department of Epidemiology, Human Genetics & Environmental Sciences, School of Public Health, University of Texas Health Science Center, Houston, Texas, United States of America
- The Brown Foundation Institute of Molecular Medicine, Research Center for Human Genetics, The University of Texas Health Science Center, Houston, Texas, United States of America
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, United States of America
| | - Thomas H. Mosley
- Gertrude C. Ford MIND Center, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
- Department of Medicine, University of Mississippi Medical Center, Jackson, Massachusetts, United States of America
| |
Collapse
|
21
|
Makitani K, Nakagawa S, Izumi Y, Akaike A, Kume T. Inhibitory effect of donepezil on bradykinin-induced increase in the intracellular calcium concentration in cultured cortical astrocytes. J Pharmacol Sci 2017; 134:37-44. [PMID: 28499726 DOI: 10.1016/j.jphs.2017.03.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 03/06/2017] [Accepted: 03/21/2017] [Indexed: 01/07/2023] Open
Abstract
Donepezil is a potent and selective acetylcholinesterase inhibitor developed for the treatment of Alzheimer's disease. In the present study, we investigated the responses of astrocytes to bradykinin, an inflammatory mediator, and the effect of donepezil on these responses using cultured cortical astrocytes. Bradykinin induced a transient increase of intracellular calcium concentration ([Ca2+]i) in cultured astrocytes. Bradykinin-induced [Ca2+]i increase was inhibited by the exposure to thapsigargin, which depletes Ca2+ stores on endoplasmic reticulum, but not by the exclusion of extracellular Ca2+. Twenty four hours pretreatment of donepezil reduced the bradykinin-induced [Ca2+]i increase. This reduction was inhibited not only by mecamylamine, a nAChR antagonist, but also by PI3K and Akt inhibitors. In addition, donepezil inhibited bradykinin-induced increase of the intracellular reactive oxygen species level in astrocytes. These results suggest that donepezil inhibits the inflammatory response induced by bradykinin via nAChR and PI3K-Akt pathway in astrocytes.
Collapse
Affiliation(s)
- Kouki Makitani
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Shota Nakagawa
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Yasuhiko Izumi
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Akinori Akaike
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan; Department of Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | - Toshiaki Kume
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan.
| |
Collapse
|
22
|
Dai Y, Zhao Y, Tomi M, Shin BC, Thamotharan S, Mazarati A, Sankar R, Wang EA, Cepeda C, Levine MS, Zhang J, Frew A, Alger JR, Clark PM, Sondhi M, Kositamongkol S, Leibovitch L, Devaskar SU. Sex-Specific Life Course Changes in the Neuro-Metabolic Phenotype of Glut3 Null Heterozygous Mice: Ketogenic Diet Ameliorates Electroencephalographic Seizures and Improves Sociability. Endocrinology 2017; 158:936-949. [PMID: 28324109 PMCID: PMC5460805 DOI: 10.1210/en.2016-1816] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 01/18/2017] [Indexed: 01/13/2023]
Abstract
We tested the hypothesis that exposure of glut3+/- mice to a ketogenic diet ameliorates autism-like features, which include aberrant behavior and electrographic seizures. We first investigated the life course sex-specific changes in basal plasma-cerebrospinal fluid (CSF)-brain metabolic profile, brain glucose transport/uptake, glucose and monocarboxylate transporter proteins, and adenosine triphosphate (ATP) in the presence or absence of systemic insulin administration. Glut3+/- male but not female mice (5 months of age) displayed reduced CSF glucose/lactate concentrations with no change in brain Glut1, Mct2, glucose uptake or ATP. Exogenous insulin-induced hypoglycemia increased brain glucose uptake in glut3+/- males alone. Higher plasma-CSF ketones (β-hydroxybutyrate) and lower brain Glut3 in females vs males proved protective in the former while enhancing vulnerability in the latter. As a consequence, increased synaptic proteins (neuroligin4 and SAPAP1) with spontaneous excitatory postsynaptic activity subsequently reduced hippocampal glucose content and increased brain amyloid β1-40 deposition in an age-dependent manner in glut3+/- males but not females (4 to 24 months of age). We then explored the protective effect of a ketogenic diet on ultrasonic vocalization, sociability, spatial learning and memory, and electroencephalogram seizures in male mice (7 days to 6 to 8 months of age) alone. A ketogenic diet partially restored sociability without affecting perturbed vocalization, spatial learning and memory, and reduced seizure events. We conclude that (1) sex-specific and age-dependent perturbations underlie the phenotype of glut3+/- mice, and (2) a ketogenic diet ameliorates seizures caused by increased cortical excitation and improves sociability, but fails to rescue vocalization and cognitive deficits in glut3+/- male mice.
Collapse
Affiliation(s)
- Yun Dai
- Department of Pediatrics, Division of Neonatology & Developmental Biology, Neonatal Research Center at the UCLA Children’s Discovery and Innovation Institute
| | - Yuanzi Zhao
- Department of Pediatrics, Division of Neonatology & Developmental Biology, Neonatal Research Center at the UCLA Children’s Discovery and Innovation Institute
| | - Masatoshi Tomi
- Department of Pediatrics, Division of Neonatology & Developmental Biology, Neonatal Research Center at the UCLA Children’s Discovery and Innovation Institute
| | - Bo-Chul Shin
- Department of Pediatrics, Division of Neonatology & Developmental Biology, Neonatal Research Center at the UCLA Children’s Discovery and Innovation Institute
| | - Shanthie Thamotharan
- Department of Pediatrics, Division of Neonatology & Developmental Biology, Neonatal Research Center at the UCLA Children’s Discovery and Innovation Institute
| | | | - Raman Sankar
- Department of Pediatrics, Division of Neurology
- Department of Neurology
| | - Elizabeth A. Wang
- Intellectual and Developmental Disabilities Research Center and Brain Research Institute
| | - Carlos Cepeda
- Intellectual and Developmental Disabilities Research Center and Brain Research Institute
| | - Michael S. Levine
- Intellectual and Developmental Disabilities Research Center and Brain Research Institute
| | - Jingjing Zhang
- Department of Neurology
- Intellectual and Developmental Disabilities Research Center and Brain Research Institute
| | - Andrew Frew
- Department of Neurology
- Ahmanson-Lovelace Brain Mapping Center
| | - Jeffry R. Alger
- Department of Neurology
- Ahmanson-Lovelace Brain Mapping Center
| | - Peter M. Clark
- Department of Molecular and Medical Pharmacology and Crump Institute for Molecular Imaging, David Geffen School of Medicine at UCLA, Los Angeles, California 90095
| | - Monica Sondhi
- Department of Pediatrics, Division of Neonatology & Developmental Biology, Neonatal Research Center at the UCLA Children’s Discovery and Innovation Institute
| | - Sudatip Kositamongkol
- Department of Pediatrics, Division of Neonatology & Developmental Biology, Neonatal Research Center at the UCLA Children’s Discovery and Innovation Institute
| | - Leah Leibovitch
- Department of Pediatrics, Division of Neonatology & Developmental Biology, Neonatal Research Center at the UCLA Children’s Discovery and Innovation Institute
| | - Sherin U. Devaskar
- Department of Pediatrics, Division of Neonatology & Developmental Biology, Neonatal Research Center at the UCLA Children’s Discovery and Innovation Institute
| |
Collapse
|
23
|
Depletion of coagulation factor XII ameliorates brain pathology and cognitive impairment in Alzheimer disease mice. Blood 2017; 129:2547-2556. [PMID: 28242605 DOI: 10.1182/blood-2016-11-753202] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 02/21/2017] [Indexed: 12/21/2022] Open
Abstract
Vascular abnormalities and inflammation are found in many Alzheimer disease (AD) patients, but whether these changes play a causative role in AD is not clear. The factor XII (FXII) -initiated contact system can trigger both vascular pathology and inflammation and is activated in AD patients and AD mice. We have investigated the role of the contact system in AD pathogenesis. Cleavage of high-molecular-weight kininogen (HK), a marker for activation of the inflammatory arm of the contact system, is increased in a mouse model of AD, and this cleavage is temporally correlated with the onset of brain inflammation. Depletion of FXII in AD mice inhibited HK cleavage in plasma and reduced neuroinflammation, fibrinogen deposition, and neurodegeneration in the brain. Moreover, FXII-depleted AD mice showed better cognitive function than untreated AD mice. These results indicate that FXII-mediated contact system activation contributes to AD pathogenesis, and therefore this system may offer novel targets for AD treatment.
Collapse
|
24
|
Bitencourt RM, Guerra de Souza AC, Bicca MA, Pamplona FA, de Mello N, Passos GF, Medeiros R, Takahashi RN, Calixto JB, Prediger RD. Blockade of hippocampal bradykinin B1 receptors improves spatial learning and memory deficits in middle-aged rats. Behav Brain Res 2017; 316:74-81. [DOI: 10.1016/j.bbr.2016.08.041] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 07/24/2016] [Accepted: 08/22/2016] [Indexed: 11/30/2022]
|
25
|
Asraf K, Torika N, Danon A, Fleisher-Berkovich S. Involvement of the Bradykinin B 1 Receptor in Microglial Activation: In Vitro and In Vivo Studies. Front Endocrinol (Lausanne) 2017; 8:82. [PMID: 28469598 PMCID: PMC5396024 DOI: 10.3389/fendo.2017.00082] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 03/30/2017] [Indexed: 11/13/2022] Open
Abstract
The importance of brain inflammation to Alzheimer's disease (AD) pathogenesis has been accepted of late, with it currently being held that brain inflammation aggravates AD pathology. One important aspect of brain inflammation is the recruitment and activation of microglia, a process termed microgliosis. Kinins and bradykinin (BK), in particular, are major pro-inflammatory mediators in the periphery, although all of the factors comprising the kinin system have also been described in the brain. Moreover, it was shown that the amyloid β (Aβ) peptide (a component of AD plaques) enhances kinin secretion and activates BK receptors that can, in turn, stimulate Aβ production. Still, the role of bradykinin in modulating brain inflammation and AD is not completely understood. In this study, we aimed to investigate the roles of the bradykinin B1 receptor (B1R) and bradykinin B2 receptor (B2R) in regulating microglial secretion of pro-inflammatory factors in vitro. Furthermore, the effects of intranasal administration of specific B1R and B2R antagonists on Aβ burden and microglial accumulation in the brains of transgenic AD mice were studied. The data obtained show that neither R-715 (a B1R antagonist) nor HOE 140 (a B2R antagonist) altered microglial cell viability. However, R-715, but not HOE 140, markedly increased lipopolysaccharide-induced nitric oxide (NO) and tumor necrosis factor-alpha (TNF-α) release, as well as inducible nitric oxide synthase expression in BV2 microglial cells. Neither antagonist altered NO nor TNF-α production in non-stimulated cells. We also showed that intranasal administration of R-715 but not HOE 140 to 8-week-old 5X familial AD mice enhanced amyloid burden and microglia/macrophage accumulation in the cortex. To conclude, we provide evidence supporting a role of B1R in brain inflammation and in the regulation of amyloid deposition in AD mice, possibly with microglial/macrophage involvement. Further studies are required to test whether modulation of this receptor can serve as a novel therapeutic strategy for AD.
Collapse
Affiliation(s)
- Keren Asraf
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Nofar Torika
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Abraham Danon
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Sigal Fleisher-Berkovich
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- *Correspondence: Sigal Fleisher-Berkovich,
| |
Collapse
|
26
|
Caetano AL, Dong-Creste KE, Amaral FA, Monteiro-Silva KC, Pesquero JB, Araujo MS, Montor WR, Viel TA, Buck HS. Kinin B2 receptor can play a neuroprotective role in Alzheimer's disease. Neuropeptides 2015; 53:51-62. [PMID: 26387425 DOI: 10.1016/j.npep.2015.09.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 09/04/2015] [Accepted: 09/05/2015] [Indexed: 12/19/2022]
Abstract
Alzheimer's disease (AD) is characterized by cognitive decline, presence of amyloid-beta peptide (Aβ) aggregates and neurofibrillary tangles. Kinins act through B1 and B2 G-protein coupled receptors (B1R and B2R). Chronic infusion of Aβ peptide leads to memory impairment and increases in densities of both kinin receptors in memory processing areas. Similar memory impairment was observed in C57BL/6 mice (WTAβ) but occurred earlier in mice lacking B2R (KOB2Aβ) and was absent in mice lacking B1R (KOB1Aβ). Thus, the aim of this study was to evaluate the participation of B1R and B2R in Aβ peptide induced cognitive deficits through the evaluation of densitiesof kinin receptors, synapses, cell bodies and number of Aβ deposits in brain ofWTAβ, KOB1Aβ and KOB2Aβ mice. An increase in B2R density was observed in both WTAβ and KOB1Aβ in memory processing related areas. KOB1Aβ showed a decrease in neuronal density and an increase in synaptic density and, in addition, an increase in Aβ deposits in KOB2Aβ was observed. In conclusion, memory preservation in KOB1Aβ, could be due to the increase in densities of B2R, suggesting a neuroprotective role for B2R, reinforced by the increased number of Aβ plaques in KOB2Aβ. Our data point to B2R as a potential therapeutic target in AD.
Collapse
Affiliation(s)
- A L Caetano
- Department of Physiological Sciences, Santa Casa de Sao Paulo School of Medical Sciences, Sao Paulo, SP CEP 01221-020, Brazil; Research Group on Neuropharmacology of Aging, Brazil
| | - K E Dong-Creste
- Department of Physiological Sciences, Santa Casa de Sao Paulo School of Medical Sciences, Sao Paulo, SP CEP 01221-020, Brazil; Research Group on Neuropharmacology of Aging, Brazil
| | - F A Amaral
- Department of Physiological Sciences, Santa Casa de Sao Paulo School of Medical Sciences, Sao Paulo, SP CEP 01221-020, Brazil
| | - K C Monteiro-Silva
- Department of Physiological Sciences, Santa Casa de Sao Paulo School of Medical Sciences, Sao Paulo, SP CEP 01221-020, Brazil
| | - J B Pesquero
- Department of Biophysics, Federal University of Sao Paulo, São Paulo, SP CEP 04021-001, Brazil
| | - M S Araujo
- Department of Biochemistry, Federal University of Sao Paulo, São Paulo, SP CEP 04021-001, Brazil
| | - W R Montor
- Department of Physiological Sciences, Santa Casa de Sao Paulo School of Medical Sciences, Sao Paulo, SP CEP 01221-020, Brazil
| | - T A Viel
- School of Arts, Sciences and Humanities and Graduation Course on Pharmacology at Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP CEP 03828-080, Brazil; Research Group on Neuropharmacology of Aging, Brazil
| | - H S Buck
- Department of Physiological Sciences, Santa Casa de Sao Paulo School of Medical Sciences, Sao Paulo, SP CEP 01221-020, Brazil; Research Group on Neuropharmacology of Aging, Brazil
| |
Collapse
|
27
|
Nascimento IC, Glaser T, Nery AA, Pillat MM, Pesquero JB, Ulrich H. Kinin-B1 and B2 receptor activity in proliferation and neural phenotype determination of mouse embryonic stem cells. Cytometry A 2015; 87:989-1000. [PMID: 26243460 DOI: 10.1002/cyto.a.22726] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The kinins bradykinin and des-arg(9) -bradykinin cleaved from kininogen precursors by kallikreins exert their biological actions by stimulating kinin-B2 and B1 receptors, respectively. In vitro models of neural differentiation such as P19 embryonal carcinoma cells and neural progenitor cells have suggested the involvement of B2 receptors in neural differentiation and phenotype determination; however, the involvement of B1 receptors in these processes has not been established. Here, we show that B1 and B2 receptors are differentially expressed in mouse embryonic E14Tg2A stem cells undergoing neural differentiation. Proliferation and differentiation assays, performed in the presence of receptor subtype-selective agonists and antagonists, revealed that B1 receptor activity is required for the proliferation of embryonic and differentiating cells as well as for neuronal maturation at later stages of differentiation, while the B2 receptor acts on neural phenotype choice, promoting neurogenesis over gliogenesis. Besides the elucidation of bradykinin functions in an in vitro model reflecting early embryogenesis and neurogenesis, this study contributes to the understanding of B1 receptor functions in this process.
Collapse
Affiliation(s)
- Isis C Nascimento
- Departamento de Neurologia e Neurocirurgia, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Talita Glaser
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Arthur A Nery
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Micheli M Pillat
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil
| | - João B Pesquero
- Departamento de Biofísica, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Henning Ulrich
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
28
|
Nguyen TP, Priami C, Caberlotto L. Novel drug target identification for the treatment of dementia using multi-relational association mining. Sci Rep 2015; 5:11104. [PMID: 26154857 PMCID: PMC4495601 DOI: 10.1038/srep11104] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 05/13/2015] [Indexed: 12/12/2022] Open
Abstract
Dementia is a neurodegenerative condition of the brain in which there is a progressive and permanent loss of cognitive and mental performance. Despite the fact that the number of people with dementia worldwide is steadily increasing and regardless of the advances in the molecular characterization of the disease, current medical treatments for dementia are purely symptomatic and hardly effective. We present a novel multi-relational association mining method that integrates the huge amount of scientific data accumulated in recent years to predict potential novel targets for innovative therapeutic treatment of dementia. Owing to the ability of processing large volumes of heterogeneous data, our method achieves a high performance and predicts numerous drug targets including several serine threonine kinase and a G-protein coupled receptor. The predicted drug targets are mainly functionally related to metabolism, cell surface receptor signaling pathways, immune response, apoptosis, and long-term memory. Among the highly represented kinase family and among the G-protein coupled receptors, DLG4 (PSD-95), and the bradikynin receptor 2 are highlighted also for their proposed role in memory and cognition, as described in previous studies. These novel putative targets hold promises for the development of novel therapeutic approaches for the treatment of dementia.
Collapse
Affiliation(s)
- Thanh-Phuong Nguyen
- The Microsoft Research, University of Trento Centre for Computational Systems Biology (COSBI), Piazza Manifattura 1, 38068, Rovereto, Italy
- Life Sciences Research Unit, University of Luxembourg, 162 A, avenue de la Faïencerie, L-1511 Luxembourg
| | - Corrado Priami
- The Microsoft Research, University of Trento Centre for Computational Systems Biology (COSBI), Piazza Manifattura 1, 38068, Rovereto, Italy
- Department of Mathematics, University of Trento, Via Sommarive, 14-38123 Povo, Italy
| | - Laura Caberlotto
- The Microsoft Research, University of Trento Centre for Computational Systems Biology (COSBI), Piazza Manifattura 1, 38068, Rovereto, Italy
| |
Collapse
|
29
|
Atorvastatin Prevents Cognitive Deficits Induced by Intracerebroventricular Amyloid-β1–40 Administration in Mice: Involvement of Glutamatergic and Antioxidant Systems. Neurotox Res 2015; 28:32-42. [DOI: 10.1007/s12640-015-9527-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 03/12/2015] [Accepted: 03/19/2015] [Indexed: 12/12/2022]
|
30
|
Bicca MA, Costa R, Loch-Neckel G, Figueiredo CP, Medeiros R, Calixto JB. B₂ receptor blockage prevents Aβ-induced cognitive impairment by neuroinflammation inhibition. Behav Brain Res 2014; 278:482-91. [PMID: 25446751 DOI: 10.1016/j.bbr.2014.10.040] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 10/23/2014] [Accepted: 10/29/2014] [Indexed: 11/28/2022]
Abstract
BACKGROUND AND PURPOSE Aβ-induced neuronal toxicity and memory loss is thought to be dependent on neuroinflammation, an important event in Alzheimer's disease (AD). Previously, we demonstrated that the blockage of the kinin B2 receptor (B2R) protects against the memory deficits induced by amyloid β (Aβ) peptide in mice. In this study, we aimed to investigate the role of B2R on Aβ-induced neuroinflammation in mice and the beneficial effects of B2R blockage in synapses alterations. EXPERIMENTAL APPROACH The selective kinin B2R antagonist HOE 140 (50 pmol/site) was given by intracerebroventricular (i.c.v.) route to male Swiss mice 2 h prior the i.c.v. injection of Aβ(1-40) (400 pmol/site) peptide. Animals were sacrificed, at specific time points after Aβ(1-40) injection (6 h, 1 day or 8 days), and the brain was collected in order to perform immunohistochemical analysis. Different groups of animals were submitted to behavioral cognition tests on day 14 after Aβ(1-40) administration. KEY RESULTS In this study, we report that the pre-treatment with the selective kinin B2R antagonist HOE 140 significantly inhibited Aβ-induced neuroinflammation in mice. B2R antagonism reduced microglial activation and the levels of pro-inflammatory proteins, including COX-2, iNOS and nNOS. Notably, these phenomena were accompanied by an inhibition of MAPKs (JNK and p38) and transcription factors (c-Jun and p65/NF-κB) activation. Finally, the anti-inflammatory effects of B2R antagonism provided significant protection against Aβ(1-40)-induced synaptic loss and cognitive impairment in mice. CONCLUSIONS AND IMPLICATIONS Collectively, these results suggest that B2R activation may play a critical role in Aβ-induced neuroinflammation, one of the most important contributors to AD progression, and its blockage can provide synapses protection.
Collapse
Affiliation(s)
- M A Bicca
- Departamento de Farmacologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, 88049-900 Florianópolis, Santa Catarina, Brazil
| | - R Costa
- Departamento de Farmacologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, 88049-900 Florianópolis, Santa Catarina, Brazil
| | - G Loch-Neckel
- Departamento de Farmacologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, 88049-900 Florianópolis, Santa Catarina, Brazil
| | - C P Figueiredo
- Departamento de Farmacologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, 88049-900 Florianópolis, Santa Catarina, Brazil
| | - R Medeiros
- Departamento de Farmacologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, 88049-900 Florianópolis, Santa Catarina, Brazil
| | - J B Calixto
- Departamento de Farmacologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, 88049-900 Florianópolis, Santa Catarina, Brazil.
| |
Collapse
|
31
|
Fournier A, Oprisiu-Fournier R, Serot JM, Godefroy O, Achard JM, Faure S, Mazouz H, Temmar M, Albu A, Bordet R, Hanon O, Gueyffier F, Wang J, Black S, Sato N. Prevention of dementia by antihypertensive drugs: how AT1-receptor-blockers and dihydropyridines better prevent dementia in hypertensive patients than thiazides and ACE-inhibitors. Expert Rev Neurother 2014; 9:1413-31. [DOI: 10.1586/ern.09.89] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
32
|
Dutra RC, Moreira ELG, Alberti TB, Marcon R, Prediger RD, Calixto JB. Spatial reference memory deficits precede motor dysfunction in an experimental autoimmune encephalomyelitis model: the role of kallikrein-kinin system. Brain Behav Immun 2013; 33:90-101. [PMID: 23777652 DOI: 10.1016/j.bbi.2013.06.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Revised: 05/21/2013] [Accepted: 06/07/2013] [Indexed: 12/28/2022] Open
Abstract
Multiple sclerosis (MS) is a progressive T cell-mediated autoimmune demyelinating inflammatory disease of the central nervous system (CNS). Although it is recognized that cognitive deficits represent a manifestation of the disease, the underlying pathogenic mechanisms remain unknown. Here we provide evidence of spatial reference memory impairments during the pre-motor phase of experimental autoimmune encephalomyelitis (EAE) in mice. Specifically, these cognitive deficits were accompanied by down-regulation of choline acetyltransferase (ChAT) mRNA expression on day 5 and 11 post-immunization, and up-regulation of inflammatory cytokines in the hippocampus and prefrontal cortex. Moreover, a marked increase in B1R mRNA expression occurred selectively in the hippocampus, whereas protein level was up-regulated in both brain areas. Genetic deletion of kinin B1R attenuated cognitive deficits and cholinergic dysfunction, and blocked mRNA expression of both IL-17 and IFN-γ in the prefrontal cortex, lymph node and spleen of mice subjected to EAE. The discovery of kinin receptors, mainly B1R, as a target for controlling neuroinflammatory response, as well as the cognitive deficits induced by EAE may foster the therapeutic exploitation of the kallikrein-kinin system (KKS), in particular for the treatment of autoimmune disorders, such as MS, mainly during pre-symptomatic phase.
Collapse
Affiliation(s)
- Rafael C Dutra
- Laboratory of Autoimmunity and Immunopharmacology, Campus Araranguá, Universidade Federal de Santa Catarina, 88900-000 Araranguá, SC, Brazil; Department of Pharmacology, Centre of Biological Sciences, Universidade Federal de Santa Catarina, 88049-900 Florianópolis, SC, Brazil.
| | | | | | | | | | | |
Collapse
|
33
|
Ma Y, Wang S, Tian Y, Chen L, Li G, Mao J. Disruption of persistent nociceptive behavior in rats with learning impairment. PLoS One 2013; 8:e74533. [PMID: 24040273 PMCID: PMC3770575 DOI: 10.1371/journal.pone.0074533] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Accepted: 08/04/2013] [Indexed: 12/02/2022] Open
Abstract
Despite the subjective nature of pain experience with cognitive and affective dimensions, preclinical pain research has largely focused on its sensory dimension. Here, we examined the relationship between learning/memory and nociceptive behavior in rats with combined learning impairment and persistent nociception. Learning impairment was induced by bilateral hippocampal injection of a mixed Aβ solution, whereas persistent nociception produced in these rats by complete Freund's adjuvant-induced ankle inflammation. Those rats with learning impairment showed a diminished development of thermal hyperalgesia and mechanical allodynia and a shorter time course of nociceptive behavior without alteration of their baseline nociceptive threshold. In rats with pre-established hyperalgesia and allodynia due to ankle inflammation, bilateral intra-hippocampal injection of cycloheximide (a protein synthesis inhibitor) promoted the earlier recovery of nociceptive behavior. Moreover, expression of Aβ, NR1 subunit of the N-methyl-D-aspartate receptor, and protein kinase Cγ was upregulated, whereas the choline acetyl transferase expression was downregulated, in the hippocampus, thalamus, amygdala, and/or spinal cord of rats with combined learning impairment and persistent nociception. The data indicate that learning impairment could disrupt the response to a state of persistent nociception, suggesting an important role for cognitive maladaptation in the mechanisms of chronic pain. These results also suggest that a preclinical model of combined learning impairment and persistent nociception may be useful to explore the brain mechanisms underlying the transition from acute to chronic pain.
Collapse
Affiliation(s)
- Yuxin Ma
- MGH Center for Translational Pain Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Anatomy, School of Basic Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Shuxing Wang
- MGH Center for Translational Pain Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Yinghong Tian
- MGH Center for Translational Pain Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Lucy Chen
- MGH Center for Translational Pain Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Guoying Li
- Department of Anatomy, School of Basic Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Jianren Mao
- MGH Center for Translational Pain Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
34
|
Blaes N, Girolami JP. Targeting the 'Janus face' of the B2-bradykinin receptor. Expert Opin Ther Targets 2013; 17:1145-66. [PMID: 23957374 DOI: 10.1517/14728222.2013.827664] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Kinins are main active mediators of the kallikrein-kinin system (KKS) via bradykinin type 1 inducible (B1R) and type 2 constitutive (B2R) receptors. B2R mediates most physiological bradykinin (BK) responses, including vasodilation, natriuresis, NO, prostaglandins release. AREAS COVERED The article summarizes knowledge on kinins, B2R signaling and biological functions; highlights crosstalks between B2R and renin-angiotensin system (RAS). The double role (Janus face) in physiopathology, namely the beneficial protection of the endothelium, which forms the basis for the therapeutical utilization of B2 receptor agonists, on the one side, and the involvement of B2R in inflammation or infection diseases and in pain mechanisms, which justifies the use of B2R antagonists, on the other side, is extensively analyzed. EXPERT OPINION For decades, the B2R has been unconsciously activated during angiotensin-converting enzyme inhibitor (ACEI) or angiotensin receptor blocker (ARB) treatments. Whether direct B2R targeting with stable agonists could bring additional therapeutic benefit to RAS inhibition should be investigated. Efficacy, established in experimental models, should be confirmed by translational studies in cardiovascular pathologies, glaucoma, Duchenne cardiopathy and during brain cancer therapy. The other face of B2R is targeted by antagonists already approved to treat hereditary angioedema. The use of antagonists could be extended to other angioedema and efficacy tested against acute pain and inflammatory diseases.
Collapse
Affiliation(s)
- Nelly Blaes
- INSERM, U1048, Institute of Metabolic and Cardiovascular Diseases, I2MC, Université Paul Sabatier , F-31432, Toulouse , France
| | | |
Collapse
|
35
|
Lacoste B, Tong XK, Lahjouji K, Couture R, Hamel E. Cognitive and cerebrovascular improvements following kinin B1 receptor blockade in Alzheimer's disease mice. J Neuroinflammation 2013; 10:57. [PMID: 23642031 PMCID: PMC3710240 DOI: 10.1186/1742-2094-10-57] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2013] [Accepted: 04/20/2013] [Indexed: 12/11/2022] Open
Abstract
Background Recent evidence suggests that the inducible kinin B1 receptor (B1R) contributes to pathogenic neuroinflammation induced by amyloid-beta (Aβ) peptide. The present study aims at identifying the cellular distribution and potentially detrimental role of B1R on cognitive and cerebrovascular functions in a mouse model of Alzheimer’s disease (AD). Methods Transgenic mice overexpressing a mutated form of the human amyloid precursor protein (APPSwe,Ind, line J20) were treated with a selective and brain penetrant B1R antagonist (SSR240612, 10 mg/kg/day for 5 or 10 weeks) or vehicle. The impact of B1R blockade was measured on i) spatial learning and memory performance in the Morris water maze, ii) cerebral blood flow (CBF) responses to sensory stimulation using laser Doppler flowmetry, and iii) reactivity of isolated cerebral arteries using online videomicroscopy. Aβ burden was quantified by ELISA and immunostaining, while other AD landmarks were measured by western blot and immunohistochemistry. Results B1R protein levels were increased in APP mouse hippocampus and, prominently, in reactive astrocytes surrounding Aβ plaques. In APP mice, B1R antagonism with SSR240612 improved spatial learning, memory and normalized protein levels of the memory-related early gene Egr-1 in the dentate gyrus of the hippocampus. B1R antagonism restored sensory-evoked CBF responses, endothelium-dependent dilations, and normalized cerebrovascular protein levels of endothelial nitric oxide synthase and B2R. In addition, SSR240612 reduced (approximately 50%) microglial, but not astroglial, activation, brain levels of soluble Aβ1-42, diffuse and dense-core Aβ plaques, and it increased protein levels of the Aβ brain efflux transporter lipoprotein receptor-related protein-1 in cerebral microvessels. Conclusion These findings show a selective upregulation of astroglial B1R in the APP mouse brain, and the capacity of the B1R antagonist to abrogate amyloidosis, cerebrovascular and memory deficits. Collectively, these findings provide convincing evidence for a role of B1R in AD pathogenesis.
Collapse
Affiliation(s)
- Baptiste Lacoste
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, 3801 University Street, Montréal, QC H3A 2B4, Canada
| | | | | | | | | |
Collapse
|
36
|
The bradykinin B1 receptor regulates Aβ deposition and neuroinflammation in Tg-SwDI mice. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 182:1740-9. [PMID: 23470163 DOI: 10.1016/j.ajpath.2013.01.021] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Revised: 11/30/2012] [Accepted: 01/08/2013] [Indexed: 02/08/2023]
Abstract
The deposition of amyloid-β peptides (Aβ) in the cerebral vasculature, a condition known as cerebral amyloid angiopathy, is increasingly recognized as an important component leading to intracerebral hemorrhage, neuroinflammation, and cognitive impairment in Alzheimer disease (AD) and related disorders. Recent studies demonstrated a role for the bradykinin B1 receptor (B1R) in cognitive deficits induced by Aβ in mice; however, its involvement in AD and cerebral amyloid angiopathy is poorly understood. Herein, we investigated the effect of B1R inhibition on AD-like neuroinflammation and amyloidosis using the transgenic mouse model (Tg-SwDI). B1R expression was found to be up-regulated in brains of Tg-SwDI mice, specifically in the vasculature, neurons, and astrocytes. Notably, administration of the B1R antagonist, R715, to 8-month-old Tg-SwDI mice for 8 weeks resulted in higher Aβ40 levels and increased thioflavin S-positive fibrillar Aβ deposition. Moreover, blockage of B1R inhibited neuroinflammation, as evidenced by the decreased accumulation of activated microglia and reactive astrocytes, diminished NF-κB activation, and reduced cytokine and chemokine levels. Together, our results indicate that B1R activation plays an important role in limiting the accumulation of Aβ in AD-like brain, likely through the regulation of activated glial cell accumulation and release of pro-inflammatory mediators. Therefore, the modulation of the receptor may represent a novel therapeutic approach for AD.
Collapse
|
37
|
dos Santos VV, Santos DB, Lach G, Rodrigues ALS, Farina M, De Lima TCM, Prediger RD. Neuropeptide Y (NPY) prevents depressive-like behavior, spatial memory deficits and oxidative stress following amyloid-β (Aβ(1-40)) administration in mice. Behav Brain Res 2013; 244:107-15. [PMID: 23396168 DOI: 10.1016/j.bbr.2013.01.039] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Revised: 01/25/2013] [Accepted: 01/28/2013] [Indexed: 12/25/2022]
Abstract
Neuropeptide Y (NPY) is a 36-amino acid peptide widely distributed in the central nervous system (CNS) that has been associated with the modulation of several functions including food intake, learning and memory, mood and neuroprotection. There is great interest in understanding the role of NPY in the deleterious effects induced by the central accumulation of amyloid-β (Aβ) peptides, a pathological hallmark of Alzheimer's disease (AD). Herein, we evaluated the effects of a single intracerebroventricular (i.c.v.) administration of NPY (0.0234 μmol/μL) 15 min prior to the i.c.v. injection of aggregated Aβ1-40 peptide (400 pmol/mouse) in behavioral and neurochemical parameters related to oxidative stress in mice. Pretreatment with NPY prevented Aβ1-40-induced depressive-like responses and spatial memory impairments evaluated in the tail suspension and object location tasks, respectively. The protective effects of NPY on spatial memory of Aβ1-40-treated mice were abolished by the pretreatment with the selective Y2 receptor antagonist BIIE0246. On the other hand, the administration of NPY and Aβ1-40 did not alter the performance of the animals in the elevated plus-maze and open field arena, indicating lack of effects on anxiety state and locomotor function. Although Aβ1-40 infusion did not change hippocampal and cortical glutathione peroxidase (GPx) activity and glutathione (GSH) levels, Aβ1-40-infused animals showed an increased lipid peroxidation in hippocampus and prefrontal cortex that were blunted by NPY administration. These findings indicate that central administration of NPY prevents Aβ1-40-induced depressive-like behavior and spatial memory deficits in mice and that this response is mediated, at least in part, by the activation of Y2 receptors and prevention of oxidative stress.
Collapse
Affiliation(s)
- Vanessa V dos Santos
- Programa de Pós-Graduação em Neurociências, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, UFSC, Florianópolis-SC, Brazil
| | | | | | | | | | | | | |
Collapse
|
38
|
Ribeiro RP, Moreira ELG, Santos DB, Colle D, Dos Santos AA, Peres KC, Figueiredo CP, Farina M. Probucol affords neuroprotection in a 6-OHDA mouse model of Parkinson's disease. Neurochem Res 2013; 38:660-8. [PMID: 23334712 DOI: 10.1007/s11064-012-0965-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Revised: 12/21/2012] [Accepted: 12/26/2012] [Indexed: 01/24/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by the degeneration of dopaminergic nigrostriatal neurons. Although the etiology of the majority of human PD cases is unknown, experimental evidence points to oxidative stress as an early and causal event. Probucol is a lipid-lowering phenolic compound with anti-inflammatory and antioxidant properties that has been recently reported as protective in neurotoxicity and neurodegeneration models. This study was designed to investigate the effects of probucol on the vulnerability of striatal dopaminergic neurons to oxidative stress in a PD in vivo model. Swiss mice were treated with probucol during 21 days (11.8 mg/kg; oral route). Two weeks after the beginning of treatment, mice received a single intracerebroventricular (i.c.v.) infusion of 6-hydroxydopamine (6-OHDA). On the 21st day, locomotor performance, striatal oxidative stress-related parameters, and striatal tyrosine hydroxylase and synaptophysin levels, were measured as outcomes of toxicity. 6-OHDA-infused mice showed hyperlocomotion and a significant decrease in striatal tyrosine hydroxylase (TH) and synaptophysin levels. In addition, 6-OHDA-infused mice showed reduced superoxide dismutase activity and increased lipid peroxidation and catalase activity in the striatum. Notably, probucol protected against 6-OHDA-induced hyperlocomotion and striatal lipid peroxidation, catalase upregulation and decrease of TH levels. Overall, the present results show that probucol protects against 6-OHDA-induced toxicity in mice. These findings may render probucol as a promising molecule for further pharmacological studies on the search for disease-modifying treatment in PD.
Collapse
Affiliation(s)
- Renata Pietsch Ribeiro
- Programa de Pós-Graduação em Neurociências, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, 88040-900, Brazil.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Therapeutic Effect of Yi-Chi-Tsung-Ming-Tang on Amyloid β-Induced Alzheimer's Disease-Like Phenotype via an Increase of Acetylcholine and Decrease of Amyloid β. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2012; 2012:414536. [PMID: 22754582 PMCID: PMC3382387 DOI: 10.1155/2012/414536] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Accepted: 04/23/2012] [Indexed: 11/17/2022]
Abstract
Alzheimer's disease (AD) is an irreversible neurodegenerative disorder characterized by amyloid accumulation, neuronal death, and cognitive impairments. Yi-Chi-Tsung-Ming-Tang (YCTMT) is a traditional Chinese medicine and has never been used to enhance cognitive function and treat neurodegenerative disorders such as senile dementia. Whether YCTMT has a beneficial role in improving learning and memory in AD patients remains unclear. The present study showed that oral administration of YCTMT ameliorated amyloid-β- (Aβ1−40) injection-induced learning and memory impairments in rats, examined using passive avoidance and Morris water-maze tests. Immunostaining and Western Blot results showed that continuous Aβ1−40 infusion caused amyloid accumulation and decreased acetylcholine level in hippocampus. Oral administration of medium and high dose of YCTMT 7 days after the Aβ1−40 infusion decreased amyloid accumulation area and reversed acetylcholine decline in the Aβ1−40-injected hippocampus, suggesting that YCTMT might inhibit Aβ plague accumulation and rescue reduced acetylcholine expression. This study has provided evidence on the beneficial role of YCTMT in ameliorating amyloid-induced AD-like symptom, indicating that YCTMT may offer an alternative strategy for treating AD.
Collapse
|
40
|
Rial D, Piermartiri TC, Duarte FS, Tasca CI, Walz R, Prediger RD. Overexpression of cellular prion protein (PrP(C)) prevents cognitive dysfunction and apoptotic neuronal cell death induced by amyloid-β (Aβ₁₋₄₀) administration in mice. Neuroscience 2012; 215:79-89. [PMID: 22537845 DOI: 10.1016/j.neuroscience.2012.04.034] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Revised: 04/02/2012] [Accepted: 04/07/2012] [Indexed: 11/28/2022]
Abstract
The cellular prion protein (PrP(C)) is a neuronal-anchored glycoprotein that has been associated with several functions in the CNS such as synaptic plasticity, learning and memory and neuroprotection. There is great interest in understanding the role of PrP(C) in the deleterious effects induced by the central accumulation of amyloid-β (Aβ) peptides, a pathological hallmark of Alzheimer's disease, but the existent results are still controversial. Here we compared the effects of a single intracerebroventricular (i.c.v.) injection of aggregated Aβ(1-40) peptide (400pmol/mouse) on the spatial learning and memory performance as well as hippocampal cell death biomarkers in adult wild type (Prnp(+/+)), PrP(C) knockout (Prnp(0/0)) and the PrP(C) overexpressing Tg-20 mice. Tg-20 mice, which present a fivefold increase in PrP(C) expression in comparison to wild type mice, were resistant to the Aβ(1-40)-induced spatial learning and memory impairments as indicated by reduced escape latencies to find the platform and higher percentage of time spent in the correct quadrant during training and probe test sessions of the water maze task. The protection against Aβ(1-40)-induced cognitive impairments observed in Tg-20 mice was accompanied by a significant decrease in the hippocampal expression of the activated caspase-3 protein and Bax/Bcl-2 ratio as well as reduced hippocampal cell damage assessed by MTT and propidium iodide incorporation assays. These findings indicate that the overexpression of PrP(C) prevents Aβ(1-40)-induced spatial learning and memory deficits in mice and that this response is mediated, at least in part, by the modulation of programed cell death pathways.
Collapse
Affiliation(s)
- D Rial
- Departamento de Farmacologia, Universidade Federal de Santa Catarina, UFSC, Florianópolis, SC, Brazil
| | | | | | | | | | | |
Collapse
|
41
|
Tacrine-6-ferulic acid, a novel multifunctional dimer, inhibits amyloid-β-mediated Alzheimer's disease-associated pathogenesis in vitro and in vivo. PLoS One 2012; 7:e31921. [PMID: 22384101 PMCID: PMC3285653 DOI: 10.1371/journal.pone.0031921] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Accepted: 01/20/2012] [Indexed: 12/13/2022] Open
Abstract
We have previously synthesized a series of hybrid compounds by linking ferulic acid to tacrine as multifunctional agents based on the hypotheses that Alzheimer's disease (AD) generates cholinergic deficiency and oxidative stress. Interestingly, we found that they may have potential pharmacological activities for treating AD. Here we report for the first time that tacrine-6-ferulic acid (T6FA), one of these compounds, can prevent amyloid-β peptide (Aβ)-induced AD-associated pathological changes in vitro and in vivo. Our results showed that T6FA significantly inhibited auto- and acetylcholinesterase (AChE)-induced aggregation of Aβ1–40in vitro and blocked the cell death induced by Aβ1–40 in PC12 cells. In an AD mouse model by the intracerebroventricular injection of Aβ1–40, T6FA significantly improved the cognitive ability along with increasing choline acetyltransferase and superoxide dismutase activity, decreasing AChE activity and malondialdehyde level. Based on our findings, we conclude that T6FA may be a promising multifunctional drug candidate for AD.
Collapse
|
42
|
Dutra RC, Leite DFP, Bento AF, Manjavachi MN, Patrício ES, Figueiredo CP, Pesquero JB, Calixto JB. The role of kinin receptors in preventing neuroinflammation and its clinical severity during experimental autoimmune encephalomyelitis in mice. PLoS One 2011; 6:e27875. [PMID: 22132157 PMCID: PMC3222659 DOI: 10.1371/journal.pone.0027875] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Accepted: 10/27/2011] [Indexed: 11/19/2022] Open
Abstract
Background Multiple sclerosis (MS) is a demyelinating and neuroinflammatory disease of the human central nervous system (CNS). The expression of kinins is increased in MS patients, but the underlying mechanisms by which the kinin receptor regulates MS development have not been elucidated. Methodology/Principal Findings Experimental autoimmune encephalomyelitis (EAE) was induced in female C57BL/6 mice by immunization with MOG35–55 peptide emulsified in complete Freund's adjuvant and injected with pertussis toxin on day 0 and day 2. Here, we report that blockade of the B1R in the induction phase of EAE markedly suppressed its progression by interfering with the onset of the immune response. Furthermore, B1R antagonist suppressed the production/expression of antigen-specific TH1 and TH17 cytokines and transcription factors, both in the periphery and in the CNS. In the chronic phase of EAE, the blockade of B1R consistently impaired the clinical progression of EAE. Conversely, administration of the B1R agonist in the acute phase of EAE suppressed disease progression and inhibited the increase in permeability of the blood-brain barrier (BBB) and any further CNS inflammation. Of note, blockade of the B2R only showed a moderate impact on all of the studied parameters of EAE progression. Conclusions/Significance Our results strongly suggest that kinin receptors, mainly the B1R subtype, play a dual role in EAE progression depending on the phase of treatment through the lymphocytes and glial cell-dependent pathways.
Collapse
MESH Headings
- Animals
- Bradykinin Receptor Antagonists
- CD4-Positive T-Lymphocytes/immunology
- Chronic Disease
- Cytokines/biosynthesis
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/complications
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Female
- Gene Deletion
- Humans
- Inflammation/complications
- Inflammation/pathology
- Inflammation/prevention & control
- Lymphoid Tissue/immunology
- Lymphoid Tissue/pathology
- Mice
- Mice, Inbred C57BL
- Models, Biological
- Myelin Sheath/metabolism
- Receptors, Bradykinin/metabolism
- Spinal Cord/metabolism
- Spinal Cord/pathology
- Th1 Cells/immunology
- Th17 Cells/immunology
Collapse
Affiliation(s)
- Rafael C. Dutra
- Department of Pharmacology, Centre of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Daniela F. P. Leite
- Department of Pharmacology, Centre of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Allisson F. Bento
- Department of Pharmacology, Centre of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Marianne N. Manjavachi
- Department of Pharmacology, Centre of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Eliziane S. Patrício
- Department of Pharmacology, Centre of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Cláudia P. Figueiredo
- Department of Pharmacology, Centre of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - João B. Pesquero
- Department of Biophysics, Universidade Federal de São Paulo, São Paulo, Brazil
| | - João B. Calixto
- Department of Pharmacology, Centre of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
- * E-mail:
| |
Collapse
|
43
|
Chambon C, Wegener N, Gravius A, Danysz W. Behavioural and cellular effects of exogenous amyloid-β peptides in rodents. Behav Brain Res 2011; 225:623-41. [PMID: 21884730 DOI: 10.1016/j.bbr.2011.08.024] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Revised: 08/10/2011] [Accepted: 08/16/2011] [Indexed: 12/29/2022]
Abstract
A better understanding of Alzheimer's disease (AD) and the development of disease modifying therapies are some of the biggest challenges of the 21st century. One of the core features of AD are amyloid plaques composed of amyloid-beta (Aβ) peptides. The first hypothesis proposed that cognitive deficits are linked to plaque-development and transgenic mice have been generated to study this link, thereby providing a good model to develop new therapeutic approaches. Since later it was recognised that in AD patients the cognitive deficit is rather correlated to soluble amyloid levels, consequently, a new hypothesis appeared associating the earliest amyloid toxicity to these soluble species. The purpose of this review is to give a summary of behavioural and cellular data obtained after soluble Aβ peptide administration into rodents' brain, thereby showing that this model is a valid tool to investigate AD pathology when no plaques are present. Additionally, this method offers an excellent, efficient model to test compounds which could act at such early stages of the disease.
Collapse
Affiliation(s)
- Caroline Chambon
- In Vivo Pharmacology, Merz Pharmaceuticals GmbH, Eckenheimer Landstrasse 100, D-60318 Frankfurt am Main, Germany.
| | | | | | | |
Collapse
|
44
|
Bicca MA, Figueiredo CP, Piermartiri TC, Meotti FC, Bouzon ZL, Tasca CI, Medeiros R, Calixto JB. The selective and competitive N-methyl-D-aspartate receptor antagonist, (-)-6-phosphonomethyl-deca-hydroisoquinoline-3-carboxylic acid, prevents synaptic toxicity induced by amyloid-β in mice. Neuroscience 2011; 192:631-41. [PMID: 21756976 DOI: 10.1016/j.neuroscience.2011.06.038] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Revised: 06/09/2011] [Accepted: 06/10/2011] [Indexed: 10/18/2022]
Abstract
The toxicity of amyloid β (Aβ) is highly associated with Alzheimer's disease (AD), which has a high incidence in elderly people worldwide. While the current treatment for moderate and severe AD includes blockage of the N-methyl-d-aspartate receptor (NMDAR), the molecular mechanisms of its effect are still poorly understood. Herein, we report that a single i.p. administration of the selective and competitive (NMDAR) antagonist LY235959 reduced Aβ neurotoxicity by preventing the down-regulation of glial glutamate transporters (glutamate-aspartate transporter (GLAST) and glutamate transporter-1 (GLT-1)), the decrease in glutamate uptake, and the production of reactive oxygen species (ROS) induced by Aβ(1-40). Importantly, the blockage of NMDAR restored the Aβ(1-40)-induced synaptic dysfunction and cognitive impairment. However, LY235959 failed to prevent the inflammatory response associated with Aβ(1-40) treatment. Altogether, our data indicate that the acute administration of Aβ promotes oxidative stress, a decrease in glutamate transporter expression, and neurotoxicity. Our results reinforce the idea that NMDAR plays a critical regulatory action in Aβ toxicity and they provide further pre-clinical evidence for the potential role of the selective and competitive NMDAR antagonists in the treatment of AD.
Collapse
Affiliation(s)
- M A Bicca
- Departamento de Farmacologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, 88049-900, Florianópolis, Santa Catarina, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Shi X, Lu X, Zhan L, Liu L, Sun M, Gong X, Sui H, Niu X, Liu S, Zheng L, Chen J, Zhou Y. Rat hippocampal proteomic alterations following intrahippocampal injection of amyloid beta peptide (1-40). Neurosci Lett 2011; 500:87-91. [PMID: 21699958 DOI: 10.1016/j.neulet.2011.06.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2010] [Revised: 05/12/2011] [Accepted: 06/04/2011] [Indexed: 12/25/2022]
Abstract
Amyloid beta peptide 1-40 (Aβ(1-40)) is closely associated with the progressive neuronal loss and cognitive decline observed in Alzheimer's disease (AD). This study aimed to establish a proteomic strategy for the profiling of AD tissues for disease-specific changes in protein abundance. Intrahippocampal injection of Aβ(1-40) induced spatial memory and learning decline in rats. Proteomic analysis revealed the changes in protein expression in the rat hippocampus treated with Aβ(1-40). Four proteins of interest which was in abundance was significantly altered in Aβ(1-40)-treated rats were identified by peptide mass fingerprint (PMF). These proteins corresponded to synapsin Ib, protein disulfide-isomerase A3 precursor, tubulin β chain and ATP synthase β subunit. Our results provide new insights into the relationship between Aβ and the pathogenesis of AD, and suggest potential targets for the therapy of AD.
Collapse
Affiliation(s)
- Xiang Shi
- Department of Traditional Chinese Medicine, The Second Affiliated Hospital, Dalian Medical University, Dalian 116023, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Cognitive-enhancing effects of polygalasaponin hydrolysate in aβ(25-35)-induced amnesic mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2011; 2011:839720. [PMID: 21423642 PMCID: PMC3057668 DOI: 10.1155/2011/839720] [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: 11/12/2010] [Accepted: 01/10/2011] [Indexed: 12/27/2022]
Abstract
Polygalasaponins are the major active constituents of Polygala tenuifolia exhibiting antiamnesic activity, but their applications are limited due to their toxicities. Evidence showed that the toxicities can be attenuated by hydrolysis. Herein, effects of a hydrolysate of polygalasaponins (HPS) on cognitive impairment induced by Aβ25−35 were assessed by Morris water maze and step-through passive avoidance tests. The impaired spatial reference memory was improved by HPS (50 and 100 mg/kg). In the acquisition trial of step-through test, HPS (50 and 100 mg/kg) increased the latency into the dark chamber and decreased the error frequency significantly (P < .05). However, no significant change was observed during the retention trial. Additionally, HPS increased the corresponding SOD activities (62.34%, 22.09%) and decreased MDA levels (28.21%, 32.35%) in both cortex and hippocampus as compared to model animals. These results show that HPS may be a useful treatment against amnesia probably via its antioxidant properties.
Collapse
|
47
|
Viana AF, Maciel IS, Dornelles FN, Figueiredo CP, Siqueira JM, Campos MM, Calixto JB. Kinin B1 receptors mediate depression-like behavior response in stressed mice treated with systemic E. coli lipopolysaccharide. J Neuroinflammation 2010; 7:98. [PMID: 21194425 PMCID: PMC3022820 DOI: 10.1186/1742-2094-7-98] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2010] [Accepted: 12/31/2010] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Kinin B1 receptors are inducible molecules up-regulated after inflammatory stimuli. This study evaluated the relevance of kinin B1 receptors in a mouse depression behavior model. METHODS Mice were exposed to a 5-min swimming session, and 30 min later they were injected with E. coli lipopolysaccharide (LPS). Depression-like behavior was assessed by determining immobility time in a tail suspension test. Different brain structures were collected for molecular and immunohistochemical studies. Anhedonia was assessed by means of a sucrose intake test. RESULTS Our protocol elicited an increase in depression-like behavior in CF1 mice, as assessed by the tail-suspension test, at 24 h. This behavior was significantly reduced by treatment with the selective B1 receptor antagonists R-715 and SSR240612. Administration of SSR240612 also prevented an increase in number of activated microglial cells in mouse hippocampus, but did not affect a reduction in expression of mRNA for brain-derived neurotrophic factor. The increased immobility time following LPS treatment was preceded by an enhancement of hippocampal and cortical B1 receptor mRNA expression (which were maximal at 1 h), and a marked production of TNFα in serum, brain and cerebrospinal fluid (between 1 and 6 h). The depression-like behavior was virtually abolished in TNFα p55 receptor-knockout mice, and increased B1 receptor mRNA expression was completely absent in this mouse strain. Furthermore, treatment with SSR240612 was also effective in preventing anhedonia in LPS-treated mice, as assessed using a sucrose preference test. CONCLUSION Our data show, for the first time, involvement of kinin B1 receptors in depressive behavioral responses, in a process likely associated with microglial activation and TNFα production. Thus, selective and orally active B1 receptor antagonists might well represent promising pharmacological tools for depression therapy.
Collapse
MESH Headings
- Animals
- Behavior, Animal/drug effects
- Behavior, Animal/physiology
- Brain/cytology
- Brain/metabolism
- Depression/physiopathology
- Humans
- Kinins
- Lipopolysaccharides/pharmacology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Microglia/cytology
- Microglia/metabolism
- Receptor, Bradykinin B1/genetics
- Receptor, Bradykinin B1/metabolism
- Receptors, Tumor Necrosis Factor, Type I/genetics
- Receptors, Tumor Necrosis Factor, Type I/metabolism
- Stress, Psychological
- Tumor Necrosis Factor Decoy Receptors/genetics
- Tumor Necrosis Factor Decoy Receptors/metabolism
- Tumor Necrosis Factor-alpha/metabolism
Collapse
Affiliation(s)
- Alice F Viana
- Pharmacology Department, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Izaque S Maciel
- Faculty of Pharmacy, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Fabiana N Dornelles
- Pharmacology Department, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Claudia P Figueiredo
- Pharmacology Department, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Jarbas M Siqueira
- Pharmacology Department, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Maria M Campos
- Faculty of Dentistry, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Institute of Toxicology, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - João B Calixto
- Pharmacology Department, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| |
Collapse
|
48
|
Atorvastatin prevents hippocampal cell death, neuroinflammation and oxidative stress following amyloid-β1–40 administration in mice: Evidence for dissociation between cognitive deficits and neuronal damage. Exp Neurol 2010; 226:274-84. [DOI: 10.1016/j.expneurol.2010.08.030] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2010] [Revised: 08/24/2010] [Accepted: 08/26/2010] [Indexed: 01/27/2023]
|
49
|
Altered emotionality leads to increased pain tolerance in amyloid β (Aβ1–40) peptide-treated mice. Behav Brain Res 2010; 212:96-102. [DOI: 10.1016/j.bbr.2010.03.052] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2010] [Revised: 03/24/2010] [Accepted: 03/29/2010] [Indexed: 11/20/2022]
|
50
|
Lemos MTR, Amaral FA, Dong KE, Bittencourt MFQP, Caetano AL, Pesquero JB, Viel TA, Buck HS. Role of kinin B1 and B2 receptors in memory consolidation during the aging process of mice. Neuropeptides 2010; 44:163-8. [PMID: 20060587 DOI: 10.1016/j.npep.2009.12.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2009] [Revised: 12/09/2009] [Accepted: 12/10/2009] [Indexed: 01/22/2023]
Abstract
Under physiological conditions, elderly people present memory deficit associated with neuronal loss. This pattern is also associated with Alzheimer's disease but, in this case, in a dramatically intensified level. Kinin receptors have been involved in neurodegeneration and increase of amyloid-beta concentration, associated with Alzheimer's disease (AD). Considering these findings, this work evaluated the role of kinin receptors in memory consolidation during the aging process. Male C57Bl/6 (wt), knock-out B1 (koB1) or B2 (koB2) mice (3, 6, 12 and 18-month-old - mo; n=10 per group) were submitted to an acquisition session, reinforcement to learning (24h later: test 1) and final test (7days later: test 2), in an active avoidance apparatus, to evaluate memory. Conditioned avoidance responses (CAR, % of 50 trials) were registered. In acquisition sessions, similar CAR were obtained among age matched animals from all strains. However, a significant decrease in CAR was observed throughout the aging process (3mo: 8.8+/-2.3%; 6mo: 4.1+/-0.6%; 12mo: 2.2+/-0.6%, 18mo: 3.6+/-0.6%, P<0.01), indicating a reduction in the learning process. In test 1, as expected, memory retention increased significantly (P<0.05) in all 3- and 6-month-old animals as well as in 12-month-old-wt and 12-month-old-koB1 (P<0.01), compared to the training session. However, 12-month-old-koB2 and all 18-month-old animals did not show an increase in memory retention. In test 2, 3- and 6-month-old wt and koB1 mice of all ages showed a significant improvement in memory (P<0.05) compared to test 1. However, 12-month-old wt and koB2 mice of all ages showed no difference in memory retention. We suggest that, during the aging process, the B1 receptor could be involved in neurodegeneration and memory loss. Nevertheless, the B2 receptor is apparently acting as a neuroprotective factor.
Collapse
Affiliation(s)
- Mayra Tolentino Resk Lemos
- Department of Physiological Sciences, Faculdade de Ciências Médicas da Santa Casa de São Paulo. Rua Dr. Cesario Motta Junior, 61, São Paulo, SP, CEP 01221-020, Brazil
| | | | | | | | | | | | | | | |
Collapse
|