1
|
Valentin-Escalera J, Leclerc M, Calon F. High-Fat Diets in Animal Models of Alzheimer's Disease: How Can Eating Too Much Fat Increase Alzheimer's Disease Risk? J Alzheimers Dis 2024; 97:977-1005. [PMID: 38217592 PMCID: PMC10836579 DOI: 10.3233/jad-230118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2023] [Indexed: 01/15/2024]
Abstract
High dietary intake of saturated fatty acids is a suspected risk factor for neurodegenerative diseases, including Alzheimer's disease (AD). To decipher the causal link behind these associations, high-fat diets (HFD) have been repeatedly investigated in animal models. Preclinical studies allow full control over dietary composition, avoiding ethical concerns in clinical trials. The goal of the present article is to provide a narrative review of reports on HFD in animal models of AD. Eligibility criteria included mouse models of AD fed a HFD defined as > 35% of fat/weight and western diets containing > 1% cholesterol or > 15% sugar. MEDLINE and Embase databases were searched from 1946 to August 2022, and 32 preclinical studies were included in the review. HFD-induced obesity and metabolic disturbances such as insulin resistance and glucose intolerance have been replicated in most studies, but with methodological variability. Most studies have found an aggravating effect of HFD on brain Aβ pathology, whereas tau pathology has been much less studied, and results are more equivocal. While most reports show HFD-induced impairment on cognitive behavior, confounding factors may blur their interpretation. In summary, despite conflicting results, exposing rodents to diets highly enriched in saturated fat induces not only metabolic defects, but also cognitive impairment often accompanied by aggravated neuropathological markers, most notably Aβ burden. Although there are important variations between methods, particularly the lack of diet characterization, these studies collectively suggest that excessive intake of saturated fat should be avoided in order to lower the incidence of AD.
Collapse
Affiliation(s)
- Josue Valentin-Escalera
- Faculté de Pharmacie, Université Laval, Québec, Canada
- Axe Neurosciences, Centre de recherche du centre Hospitalier de l’Université Laval (CHUL), Québec, Canada
- Institut sur la Nutrition et les Aliments Fonctionnels, Québec, Canada
- OptiNutriBrain – Laboratoire International Associé (NutriNeuro France-INAF Canada)
| | - Manon Leclerc
- Faculté de Pharmacie, Université Laval, Québec, Canada
- Axe Neurosciences, Centre de recherche du centre Hospitalier de l’Université Laval (CHUL), Québec, Canada
- Institut sur la Nutrition et les Aliments Fonctionnels, Québec, Canada
- OptiNutriBrain – Laboratoire International Associé (NutriNeuro France-INAF Canada)
| | - Frédéric Calon
- Faculté de Pharmacie, Université Laval, Québec, Canada
- Axe Neurosciences, Centre de recherche du centre Hospitalier de l’Université Laval (CHUL), Québec, Canada
- Institut sur la Nutrition et les Aliments Fonctionnels, Québec, Canada
- OptiNutriBrain – Laboratoire International Associé (NutriNeuro France-INAF Canada)
| |
Collapse
|
2
|
Gorina YV, Vlasova OL, Bolshakova AV, Salmina AB. Alzheimer’s Disease: a Search for the Best Experimental Models to Decode Cellular and Molecular Mechanisms of Its Development. J EVOL BIOCHEM PHYS+ 2023. [DOI: 10.1134/s0022093023010106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2023]
|
3
|
Rab35 and glucocorticoids regulate APP and BACE1 trafficking to modulate Aβ production. Cell Death Dis 2021; 12:1137. [PMID: 34876559 PMCID: PMC8651661 DOI: 10.1038/s41419-021-04433-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 11/08/2021] [Accepted: 11/23/2021] [Indexed: 11/08/2022]
Abstract
Chronic stress and elevated glucocorticoids (GCs), the major stress hormones, are risk factors for Alzheimer’s disease (AD) and promote AD pathomechanisms, including overproduction of toxic amyloid-β (Aβ) peptides and intraneuronal accumulation of hyperphosphorylated Tau protein. The latter is linked to downregulation of the small GTPase Rab35, which mediates Tau degradation via the endolysosomal pathway. Whether Rab35 is also involved in Aβ overproduction remains an open question. Here, we find that hippocampal Rab35 levels are decreased not only by stress/GC but also by aging, another AD risk factor. Moreover, we show that Rab35 negatively regulates Aβ production by sorting amyloid precursor protein (APP) and β-secretase (BACE1) out of the endosomal network, where they interact to produce Aβ. Interestingly, Rab35 coordinates distinct intracellular trafficking steps for BACE1 and APP, mediated by its effectors OCRL and ACAP2, respectively. Finally, we demonstrate that Rab35 overexpression prevents the amyloidogenic trafficking of APP and BACE1 induced by high GC levels. These studies identify Rab35 as a key regulator of APP processing and suggest that its downregulation may contribute to stress-related and AD-related amyloidogenesis.
Collapse
|
4
|
Sharma HS, Muresanu DF, Castellani RJ, Nozari A, Lafuente JV, Buzoianu AD, Sahib S, Tian ZR, Bryukhovetskiy I, Manzhulo I, Menon PK, Patnaik R, Wiklund L, Sharma A. Alzheimer's disease neuropathology is exacerbated following traumatic brain injury. Neuroprotection by co-administration of nanowired mesenchymal stem cells and cerebrolysin with monoclonal antibodies to amyloid beta peptide. PROGRESS IN BRAIN RESEARCH 2021; 265:1-97. [PMID: 34560919 DOI: 10.1016/bs.pbr.2021.04.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Military personnel are prone to traumatic brain injury (TBI) that is one of the risk factors in developing Alzheimer's disease (AD) at a later stage. TBI induces breakdown of the blood-brain barrier (BBB) to serum proteins into the brain and leads to extravasation of plasma amyloid beta peptide (ΑβP) into the brain fluid compartments causing AD brain pathology. Thus, there is a need to expand our knowledge on the role of TBI in AD. In addition, exploration of the novel roles of nanomedicine in AD and TBI for neuroprotection is the need of the hour. Since stem cells and neurotrophic factors play important roles in TBI and in AD, it is likely that nanodelivery of these agents exert superior neuroprotection in TBI induced exacerbation of AD brain pathology. In this review, these aspects are examined in details based on our own investigations in the light of current scientific literature in the field. Our observations show that TBI exacerbates AD brain pathology and TiO2 nanowired delivery of mesenchymal stem cells together with cerebrolysin-a balanced composition of several neurotrophic factors and active peptide fragments, and monoclonal antibodies to amyloid beta protein thwarted the development of neuropathology following TBI in AD, not reported earlier.
Collapse
Affiliation(s)
- Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| | - Dafin F Muresanu
- Department of Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania; "RoNeuro" Institute for Neurological Research and Diagnostic, Cluj-Napoca, Romania
| | - Rudy J Castellani
- Department of Pathology, University of Maryland, Baltimore, MD, United States
| | - Ala Nozari
- Anesthesiology & Intensive Care, Massachusetts General Hospital, Boston, MA, United States
| | - José Vicente Lafuente
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Anca D Buzoianu
- Department of Clinical Pharmacology and Toxicology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Seaab Sahib
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - Z Ryan Tian
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - Igor Bryukhovetskiy
- Department of Fundamental Medicine, School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia; Laboratory of Pharmacology, National Scientific Center of Marine Biology, Far East Branch of the Russian Academy of Sciences, Vladivostok, Russia
| | - Igor Manzhulo
- Department of Fundamental Medicine, School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia; Laboratory of Pharmacology, National Scientific Center of Marine Biology, Far East Branch of the Russian Academy of Sciences, Vladivostok, Russia
| | - Preeti K Menon
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Ranjana Patnaik
- Department of Biomaterials, School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi, India
| | - Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
5
|
Zaretsky DV, Zaretskaia MV. Mini-review: Amyloid degradation toxicity hypothesis of Alzheimer's disease. Neurosci Lett 2021; 756:135959. [PMID: 34000347 DOI: 10.1016/j.neulet.2021.135959] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 04/28/2021] [Accepted: 05/10/2021] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia affecting millions of people. Neuronal death in AD is initiated by oligomeric amyloid-β (Aβ) peptides. The amyloid channel hypothesis readily explains the primary molecular damage but does not address major observations associated with AD such as autophagy failure and decreased metabolism. The amyloid degradation toxicity hypothesis provides the interpretation as a sequence of molecular events. Aβ enters a cell by endocytosis, and the endocytic vesicle is merged with a lysosome. Lysosomal peptidases degrade the peptide. Fragments form membrane channels in lysosomal membranes that have a significant negative charge due to the presence of acidic phospholipids. Amyloid channels can transfer various ions (including protons) and even relatively large compounds, which explains lysosomal permeabilization. The neutralization of lysosomal content inactivates degradation enzymes, results in an accumulation of undigested amyloid, and stalls autophagy. Inadequate quality control of mitochondria is associated with an increased production of reactive oxygen species and decreased energy production. Also, the passage of lysosomal proteases through rare extremely large channels results in cell death. Proposed hypothesis identifies biochemical pathways involved in the initiation and progression of cellular damage induced by beta-amyloid and provides new potential pharmacological targets to treat Alzheimer's disease.
Collapse
|
6
|
Liu J, Zuo X, Han J, Dai Q, Xu H, Liu Y, Cui S. MiR-9-5p inhibits mitochondrial damage and oxidative stress in AD cell models by targeting GSK-3β. Biosci Biotechnol Biochem 2020; 84:2273-2280. [PMID: 32713252 DOI: 10.1080/09168451.2020.1797469] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
This study aims to investigate the effects and underlying mechanisms of overexpression microRNA-9-5p (miR-9-5p) on the Aβ-induced mouse hippocampal neuron cell line HT22. Different concentrations of Aβ25-35 (10, 20, 40, 80, and 160 μM) treatment were used to establish AD model in HT22 cells. The CCK-8 assay was used to measure the cell viability. The mRNA expression levels of miR-9-5p and glycogen synthase kinase-3β (GSK-3β) were determined by RT-qPCR. HT22 cell apoptosis was analyzed flow cytometry. MiR-9-5p was down-regulated in Aβ25-35-induced HT22 cells. GSK-3β is a functional target for miR-9-5p. MiR-9-5p overexpression inhibited Aβ25-35-induced mitochondrial dysfunction, cell apoptosis, and oxidative stress by regulating GSK-3β expression in HT22 cells. Furthermore, through targeting GSK-3β, overexpression of miR-9-5p partly activated nuclear factor Nrf2/Keap1 signaling, including part increases of Nrf2, HO-1, SOD-1, GCLC expression and slight decrease of Keap1 expression. Our results showed miR-9-5p may play a powerful role in the pathogenesis of AD.
Collapse
Affiliation(s)
- Junli Liu
- Department of Geriatrics, Affiliated Hospital of Qinghai University , Xining, China
| | - Xiaoqin Zuo
- Department of Geriatrics, Affiliated Hospital of Qinghai University , Xining, China
| | - Jixiang Han
- Department of Geriatrics, Affiliated Hospital of Qinghai University , Xining, China
| | - Qingxiang Dai
- Department of Geriatrics, Affiliated Hospital of Qinghai University , Xining, China
| | - Huining Xu
- Department of Geriatrics, Affiliated Hospital of Qinghai University , Xining, China
| | - Ying Liu
- Department of Geriatrics, Affiliated Hospital of Qinghai University , Xining, China
| | - Sen Cui
- Department of Hematology, Affiliated Hospital of Qinghai University , Xining, China
| |
Collapse
|
7
|
McAllister BB, Lacoursiere SG, Sutherland RJ, Mohajerani MH. Intracerebral seeding of amyloid-β and tau pathology in mice: Factors underlying prion-like spreading and comparisons with α-synuclein. Neurosci Biobehav Rev 2020; 112:1-27. [PMID: 31996301 DOI: 10.1016/j.neubiorev.2020.01.026] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 01/15/2020] [Accepted: 01/21/2020] [Indexed: 01/08/2023]
Abstract
Alzheimer's disease (AD) is characterized neuropathologically by progressive neurodegeneration and by the presence of amyloid plaques and neurofibrillary tangles. These plaques and tangles are composed, respectively, of amyloid-beta (Aβ) and tau proteins. While long recognized as hallmarks of AD, it remains unclear what causes the formation of these insoluble deposits. One theory holds that prion-like templated misfolding of Aβ and tau induces these proteins to form pathological aggregates, and propagation of this misfolding causes the stereotyped progression of pathology commonly seen in AD. Supporting this theory, numerous studies have been conducted in which aggregated Aβ, tau, or α-synuclein is injected intracerebrally into pathology-free host animals, resulting in robust formation of pathology. Here, we review this literature, focusing on in vivo intracerebral seeding of Aβ and tau in mice. We compare the results of these experiments to what is known about the seeding and spread of α-synuclein pathology, and we discuss how this research informs our understanding of the factors underlying the onset, progression, and outcomes of proteinaceous pathologies.
Collapse
Affiliation(s)
- Brendan B McAllister
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, AB, T1K 3M4, Canada
| | - Sean G Lacoursiere
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, AB, T1K 3M4, Canada
| | - Robert J Sutherland
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, AB, T1K 3M4, Canada.
| | - Majid H Mohajerani
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, AB, T1K 3M4, Canada.
| |
Collapse
|
8
|
Commins S, Kirby BP. The complexities of behavioural assessment in neurodegenerative disorders: A focus on Alzheimer’s disease. Pharmacol Res 2019; 147:104363. [DOI: 10.1016/j.phrs.2019.104363] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/12/2019] [Accepted: 07/19/2019] [Indexed: 01/21/2023]
|
9
|
Patricio-Martínez A, Sánchez-Zavaleta R, Angulo-Cruz I, Gutierrez-Praxedis L, Ramírez E, Martínez-García I, Limón ID. The Acute Activation of the CB1 Receptor in the Hippocampus Decreases Neurotoxicity and Prevents Spatial Memory Impairment in Rats Lesioned with β-Amyloid 25-35. Neuroscience 2019; 416:239-254. [PMID: 31400487 DOI: 10.1016/j.neuroscience.2019.08.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 07/31/2019] [Accepted: 08/01/2019] [Indexed: 12/23/2022]
Abstract
Given their anti-inflammatory properties, cannabinoids have been shown to be neuroprotective agents and to reduce excitotoxicity, through the activation of the Cannabinoid receptor type 1 (CB1r). These properties have led to CB1r being proposed as pharmacological targets for the treatment of various neurodegenerative diseases. Amyloid-β 25-35 (Aβ25-35) induces the expression of inducible nitric oxide synthase (iNOS) and increases nitric oxide (NO●) levels. It has been observed that increased NO● concentrations trigger biochemical pathways that contribute to neuronal death and cognitive damage. This study aimed to evaluate the neuroprotective effect of an acute activation of CB1r on spatial memory and its impact on iNOS protein expression, NO● levels, gliosis and the neurodegenerative process induced by the injection of Aβ(25-35) into the CA1 subfield of the hippocampus. ACEA [1 μM/1 μL] and Aβ(25-35) [100 μM/1 μL] and their respective vehicle groups were injected into the CA1 subfield of the hippocampus. The animals were tested for spatial learning and memory in the eight-arm radial maze, with the results revealing that the administration of ACEA plus Aβ(25-35) improves learning and memory processes, in contrast with the Aβ(25-35) group. Moreover, ACEA plus Aβ(25-35) prevented both the increase in iNOS protein and NO● levels and the reactive gliosis induced by Aβ(25-35). Importantly, neurodegeneration was significantly reduced by the administration of ACEA plus Aβ(25-35) in the CA1 subfield of the hippocampus. The data obtained in the present research suggest that the acute early activation of CB1r is crucial for neuroprotection.
Collapse
Affiliation(s)
- Aleidy Patricio-Martínez
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas-Benemérita Universidad Autónoma de Puebla, Puebla, Mexico; Facultad de Ciencias Biológicas-Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Rodolfo Sánchez-Zavaleta
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas-Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Isael Angulo-Cruz
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas-Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Liliana Gutierrez-Praxedis
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas-Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Eleazar Ramírez
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas-Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Isabel Martínez-García
- Laboratorio de Neuroquímica, Facultad de Ciencias Químicas-Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Ilhuicamina Daniel Limón
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas-Benemérita Universidad Autónoma de Puebla, Puebla, Mexico.
| |
Collapse
|
10
|
Effects of Eugenol on Alzheimer’s Disease-like Manifestations in Insulin- and Aβ-Induced Rat Models. NEUROPHYSIOLOGY+ 2019. [DOI: 10.1007/s11062-019-09801-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
11
|
Govindpani K, McNamara LG, Smith NR, Vinnakota C, Waldvogel HJ, Faull RL, Kwakowsky A. Vascular Dysfunction in Alzheimer's Disease: A Prelude to the Pathological Process or a Consequence of It? J Clin Med 2019; 8:E651. [PMID: 31083442 PMCID: PMC6571853 DOI: 10.3390/jcm8050651] [Citation(s) in RCA: 154] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 04/29/2019] [Accepted: 05/06/2019] [Indexed: 12/19/2022] Open
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia. Despite decades of research following several theoretical and clinical lines, all existing treatments for the disorder are purely symptomatic. AD research has traditionally been focused on neuronal and glial dysfunction. Although there is a wealth of evidence pointing to a significant vascular component in the disease, this angle has been relatively poorly explored. In this review, we consider the various aspects of vascular dysfunction in AD, which has a significant impact on brain metabolism and homeostasis and the clearance of β-amyloid and other toxic metabolites. This may potentially precede the onset of the hallmark pathophysiological and cognitive symptoms of the disease. Pathological changes in vessel haemodynamics, angiogenesis, vascular cell function, vascular coverage, blood-brain barrier permeability and immune cell migration may be related to amyloid toxicity, oxidative stress and apolipoprotein E (APOE) genotype. These vascular deficits may in turn contribute to parenchymal amyloid deposition, neurotoxicity, glial activation and metabolic dysfunction in multiple cell types. A vicious feedback cycle ensues, with progressively worsening neuronal and vascular pathology through the course of the disease. Thus, a better appreciation for the importance of vascular dysfunction in AD may open new avenues for research and therapy.
Collapse
Affiliation(s)
- Karan Govindpani
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| | - Laura G McNamara
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| | - Nicholas R Smith
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| | - Chitra Vinnakota
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| | - Henry J Waldvogel
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| | - Richard Lm Faull
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| | - Andrea Kwakowsky
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
12
|
Chronic Amyloid β Oligomer Infusion Evokes Sustained Inflammation and Microglial Changes in the Rat Hippocampus via NLRP3. Neuroscience 2019. [DOI: 10.1016/j.neuroscience.2018.02.046] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
13
|
Kelly ÁM. Exercise-Induced Modulation of Neuroinflammation in Models of Alzheimer's Disease. Brain Plast 2018; 4:81-94. [PMID: 30564548 PMCID: PMC6296260 DOI: 10.3233/bpl-180074] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/11/2018] [Indexed: 12/17/2022] Open
Abstract
Alzheimer's disease (AD), a progressive, neurodegenerative condition characterised by accumulation of toxic βeta-amyloid (Aβ) plaques, is one of the leading causes of dementia globally. The cognitive impairment that is a hallmark of AD may be caused by inflammation in the brain triggered and maintained by the presence of Aβ protein, ultimately leading to neuronal dysfunction and loss. Since there is a significant inflammatory component to AD, it is postulated that anti-inflammatory strategies may be of prophylactic or therapeutic benefit in AD. One such strategy is that of regular physical activity, which has been shown in epidemiological studies to be protective against various forms of dementia including AD. Exercise induces an anti-inflammatory environment in peripheral organs and also increases expression of anti-inflammatory molecules within the brain. Here we review the evidence, mainly from animal models of AD, supporting the hypothesis that exercise can reduce or slow the cellular and cognitive impairments associated with AD by modulating neuroinflammation.
Collapse
Affiliation(s)
- Áine M. Kelly
- Department of Physiology, School of Medicine & Trinity College Institute of Neuroscience & Trinity Biomedical Sciences Institute, Trinity College Dublin, University of Dublin, Dublin, Ireland
| |
Collapse
|
14
|
Tayler HM, Palmer JC, Thomas TL, Kehoe PG, Paton JF, Love S. Cerebral Aβ 40 and systemic hypertension. J Cereb Blood Flow Metab 2018; 38:1993-2005. [PMID: 28782443 PMCID: PMC6259324 DOI: 10.1177/0271678x17724930] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mid-life hypertension and cerebral hypoperfusion may be preclinical abnormalities in people who later develop Alzheimer's disease. Although accumulation of amyloid-beta (Aβ) is characteristic of Alzheimer's disease and is associated with upregulation of the vasoconstrictor peptide endothelin-1 within the brain, it is unclear how this affects systemic arterial pressure. We have investigated whether infusion of Aβ40 into ventricular cerebrospinal fluid modulates blood pressure in the Dahl salt-sensitive rat. The Dahl salt-sensitive rat develops hypertension if given a high-salt diet. Intracerebroventricular infusion of Aβ induced a progressive rise in blood pressure in rats with pre-existing hypertension produced by a high-salt diet ( p < 0.0001), but no change in blood pressure in normotensive rats. The elevation in arterial pressure in high-salt rats was associated with an increase in low frequency spectral density in systolic blood pressure, suggesting autonomic imbalance, and reduced cardiac baroreflex gain. Our results demonstrate the potential for intracerebral Aβ to exacerbate hypertension, through modulation of autonomic activity. Present findings raise the possibility that mid-life hypertension in people who subsequently develop Alzheimer's disease may in some cases be a physiological response to reduced cerebral perfusion complicating the accumulation of Aβ within the brain.
Collapse
Affiliation(s)
- Hannah M Tayler
- 1 School of Clinical Sciences, University of Bristol, Bristol, UK
| | | | - Taya L Thomas
- 1 School of Clinical Sciences, University of Bristol, Bristol, UK
| | - Patrick G Kehoe
- 1 School of Clinical Sciences, University of Bristol, Bristol, UK
| | - Julian Fr Paton
- 2 School of Physiology, Pharmacology & Neuroscience, Biomedical Sciences, University of Bristol, Bristol, UK
| | - Seth Love
- 1 School of Clinical Sciences, University of Bristol, Bristol, UK
| |
Collapse
|
15
|
Macêdo PT, Aquino ACQ, Meurer YSR, Brandão LEM, Campêlo CLC, Lima RH, Costa MR, Ribeiro AM, Silva RH. Subtle Alterations in Spatial Memory Induced by Amyloid Peptides Infusion in Rats. Front Aging Neurosci 2018; 10:18. [PMID: 29441014 PMCID: PMC5797637 DOI: 10.3389/fnagi.2018.00018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 01/15/2018] [Indexed: 12/20/2022] Open
Abstract
The cause of Alzheimer's disease (AD) remains uncertain. The accumulation of amyloid peptides (Aβ) is the main pathophysiological hallmark of the disease. Spatial deficit is an important initial sign of AD, while other types of memory impairments that appear in later stages. The Barnes maze allows the detection of subtle alterations in spatial search by the analysis of use of different strategies. Previous findings showed a general performance deficit in this task following long-term (35 days) infusion of Aβ, which corresponds to the moderate or severe impairments of the disease. In the present study, we evaluated the effects of a low-dose 15-day long treatment with Aβ peptides on spatial and non-spatial strategies of rats tested in the Barnes maze. Aβ peptides (0.5 μL/site/day; 30 pmoL solution of Aβ1-40:Aβ1-42 10:1) or saline were bilaterally infused into the CA1 (on the first treatment day) and intraventricularly (on the following 15 days) in 6-month-old Wistar male rats. Aβ infusion induced a deficit in the performance (increased latency and distance traveled to reach the target compared to saline group). In addition, a significant association between treatment and search strategy in the retrieval trial was found: Aβ group preferred the non-spatial search strategy, while saline group preferred the spatial search. In conclusion, the protocol of Aβ infusion used here induced a subtle cognitive deficit that was specific to spatial aspects. Indeed, animals under Aβ treatment still showed retrieval, but using non-spatial strategies. We suggest that this approach is potentially useful to the study of the initial memory deficits in early AD.
Collapse
Affiliation(s)
- Priscila Tavares Macêdo
- Memory Studies Laboratory, Physiology Department, Universidade Federal do Rio Grande do Norte, Natal, Brazil.,Brain Institute, Universidade Federal do Rio Grande do Norte, Natal, Brazil
| | - Antônio C Q Aquino
- Memory Studies Laboratory, Physiology Department, Universidade Federal do Rio Grande do Norte, Natal, Brazil
| | - Ywlliane S R Meurer
- Memory Studies Laboratory, Physiology Department, Universidade Federal do Rio Grande do Norte, Natal, Brazil.,Behavioral Neuroscience Laboratory, Pharmacology Department, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Luiz E M Brandão
- Memory Studies Laboratory, Physiology Department, Universidade Federal do Rio Grande do Norte, Natal, Brazil
| | - Clarissa L C Campêlo
- Memory Studies Laboratory, Physiology Department, Universidade Federal do Rio Grande do Norte, Natal, Brazil
| | - Ramon H Lima
- Memory Studies Laboratory, Physiology Department, Universidade Federal do Rio Grande do Norte, Natal, Brazil
| | - Marcos R Costa
- Brain Institute, Universidade Federal do Rio Grande do Norte, Natal, Brazil
| | - Alessandra M Ribeiro
- Laboratory of Neuroscience and Bioprospecting of Natural Products, Department of Biosciences, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Regina H Silva
- Behavioral Neuroscience Laboratory, Pharmacology Department, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
16
|
Nixon RA. Amyloid precursor protein and endosomal-lysosomal dysfunction in Alzheimer's disease: inseparable partners in a multifactorial disease. FASEB J 2017; 31:2729-2743. [PMID: 28663518 DOI: 10.1096/fj.201700359] [Citation(s) in RCA: 238] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 04/21/2017] [Indexed: 12/15/2022]
Abstract
Abnormalities of the endosomal-lysosomal network (ELN) are a signature feature of Alzheimer's disease (AD). These include the earliest known cytopathology that is specific to AD and that affects endosomes and induces the progressive failure of lysosomes, each of which are directly linked by distinct mechanisms to neurodegeneration. The origins of ELN dysfunction and β-amyloidogenesis closely overlap, which reflects their common genetic basis, the established early involvement of endosomes and lysosomes in amyloid precursor protein (APP) processing and clearance, and the pathologic effect of certain APP metabolites on ELN functions. Genes that promote β-amyloidogenesis in AD (APP, PSEN1/2, and APOE4) have primary effects on ELN function. The importance of primary ELN dysfunction to pathogenesis is underscored by the mutations in more than 35 ELN-related genes that, thus far, are known to cause familial neurodegenerative diseases even though different pathogenic proteins may be involved. In this article, I discuss growing evidence that implicates AD gene-driven ELN disruptions as not only the antecedent pathobiology that underlies β-amyloidogenesis but also as the essential partner with APP and its metabolites that drive the development of AD, including tauopathy, synaptic dysfunction, and neurodegeneration. The striking amelioration of diverse deficits in animal AD models by remediating ELN dysfunction further supports a need to integrate APP and ELN relationships, including the role of amyloid-β, into a broader conceptual framework of how AD arises, progresses, and may be effectively therapeutically targeted.-Nixon, R. A. Amyloid precursor protein and endosomal-lysosomal dysfunction in Alzheimer's disease: inseparable partners in a multifactorial disease.
Collapse
Affiliation(s)
- Ralph A Nixon
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, New York, USA; .,Department of Psychiatry and Department of Cell Biology, New York University Langone Medical Center, New York, New York, USA
| |
Collapse
|
17
|
Mateen BA, Hill CS, Biddie SC, Menon DK. DNA Methylation: Basic Biology and Application to Traumatic Brain Injury. J Neurotrauma 2017; 34:2379-2388. [DOI: 10.1089/neu.2017.5007] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Affiliation(s)
- Bilal A. Mateen
- Division of Medicine, University College London, London, United Kingdom
| | - Ciaran S. Hill
- John van Geest Centre for Brain Repair, School of Clinical Medicine, Addenbrookes Hospital, Cambridge, United Kingdom
| | - Simon C. Biddie
- Division of Anaesthesia, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - David K. Menon
- John van Geest Centre for Brain Repair, School of Clinical Medicine, Addenbrookes Hospital, Cambridge, United Kingdom
- Division of Anaesthesia, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
- Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
18
|
Sagy-Bross C, Kasianov K, Solomonov Y, Braiman A, Friedman A, Hadad N, Levy R. The role of cytosolic phospholipase A2 α in amyloid precursor protein induction by amyloid beta1-42 : implication for neurodegeneration. J Neurochem 2015; 132:559-71. [PMID: 25533654 DOI: 10.1111/jnc.13012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 12/02/2014] [Accepted: 12/03/2014] [Indexed: 01/15/2023]
Abstract
Amyloid-β peptides generated by proteolysis of the β-amyloid precursor protein (APP) play an important role in the pathogenesis of Alzheimer's disease. The present study aimed to determine whether cytosolic phospholipase A2 α (cPLA2 α) plays a role in elevated APP protein expression induced by aggregated amyloid-β1-42 (Aβ) in cortical neurons and to elucidate its specific role in signal events leading to APP induction. Elevated cPLA2 α and its activity determined by phosphorylation on serine 505 as well as elevated APP protein expression, were detected in primary rat cortical neuronal cultures exposed to Aβ for 24 h and in cortical neuron of human amyloid-β1-42 brain infused mice. Prevention of cPLA2 α up-regulation and its activity by oligonucleotide antisense against cPLA2 α (AS) prevented the elevation of APP protein in cortical neuronal cultures and in mouse neuronal cortex. To determine the role of cPLA2 α in the signals leading to APP induction, increased cPLA2 α expression and activity induced by Aβ was prevented by means of AS in neuronal cortical cultures. Under these conditions, the elevated cyclooxygenase-2 and the production of prostaglandin E2 (PGE2 ) were prevented. Addition of PGE2 or cyclic AMP analogue (dbcAMP) to neuronal cultures significantly increased the expression of APP protein, while the presence protein kinase A inhibitor (H-89) attenuated the elevation of APP induced by Aβ. Inhibition of elevated cPLA2 α by AS prevented the activation of cAMP response element binding protein (CREB) as detected by its phosphorylated form, its translocation to the nucleus and its DNA binding induced by Aβ which coincided with cPLA2 α dependent activation of CREB in the cortex of Aβ brain infused mice. Our results show that accumulation of Aβ induced elevation of APP protein expression mediated by cPLA2 α, PGE2 release, and CREB activation via protein kinase A pathway.
Collapse
Affiliation(s)
- Chen Sagy-Bross
- Immunology and Infectious Diseases Laboratory, Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev and Soroka University Medical Center, Beer-Sheva, Israel
| | - Ksenia Kasianov
- Immunology and Infectious Diseases Laboratory, Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev and Soroka University Medical Center, Beer-Sheva, Israel
| | - Yulia Solomonov
- Immunology and Infectious Diseases Laboratory, Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev and Soroka University Medical Center, Beer-Sheva, Israel
| | - Alex Braiman
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Alon Friedman
- Department of Physiology and cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Nurit Hadad
- Immunology and Infectious Diseases Laboratory, Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev and Soroka University Medical Center, Beer-Sheva, Israel
| | - Rachel Levy
- Immunology and Infectious Diseases Laboratory, Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev and Soroka University Medical Center, Beer-Sheva, Israel
| |
Collapse
|
19
|
Ribeiro FM, Camargos ERDS, de Souza LC, Teixeira AL. Animal models of neurodegenerative diseases. BRAZILIAN JOURNAL OF PSYCHIATRY 2014; 35 Suppl 2:S82-91. [PMID: 24271230 DOI: 10.1590/1516-4446-2013-1157] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The prevalence of neurodegenerative diseases, such as Alzheimer's disease (AD) and Parkinson's disease (PD), increases with age, and the number of affected patients is expected to increase worldwide in the next decades. Accurately understanding the etiopathogenic mechanisms of these diseases is a crucial step for developing disease-modifying drugs able to preclude their emergence or at least slow their progression. Animal models contribute to increase the knowledge on the pathophysiology of neurodegenerative diseases. These models reproduce different aspects of a given disease, as well as the histopathological lesions and its main symptoms. The purpose of this review is to present the main animal models for AD, PD, and Huntington's disease.
Collapse
Affiliation(s)
- Fabíola Mara Ribeiro
- Neurobiochemistry Laboratory, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo HorizonteMG, Brazil
| | | | | | | |
Collapse
|
20
|
McLarnon JG. Correlated inflammatory responses and neurodegeneration in peptide-injected animal models of Alzheimer's disease. BIOMED RESEARCH INTERNATIONAL 2014; 2014:923670. [PMID: 24822221 PMCID: PMC4005142 DOI: 10.1155/2014/923670] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 02/26/2014] [Accepted: 02/27/2014] [Indexed: 12/15/2022]
Abstract
Animal models of Alzheimer's disease (AD) which emphasize activation of microglia may have particular utility in correlating proinflammatory activity with neurodegeneration. This paper reviews injection of amyloid- β (A β ) into rat brain as an alternative AD animal model to the use of transgenic animals. In particular, intrahippocampal injection of Aβ 1-42 peptide demonstrates prominent microglial mobilization and activation accompanied by a significant loss of granule cell neurons. Furthermore, pharmacological inhibition of inflammatory reactivity is demonstrated by a broad spectrum of drugs with a common endpoint in conferring neuroprotection in peptide-injected animals. Peptide-injection models provide a focus on glial cell responses to direct peptide injection in rat brain and offer advantages in the study of the mechanisms underlying neuroinflammation in AD brain.
Collapse
Affiliation(s)
- James G. McLarnon
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC, Canada V6T 1W3
| |
Collapse
|
21
|
Alvarado-Martínez R, Salgado-Puga K, Peña-Ortega F. Amyloid beta inhibits olfactory bulb activity and the ability to smell. PLoS One 2013; 8:e75745. [PMID: 24086624 PMCID: PMC3784413 DOI: 10.1371/journal.pone.0075745] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Accepted: 08/20/2013] [Indexed: 11/17/2022] Open
Abstract
Early olfactory dysfunction has been consistently reported in both Alzheimer's disease (AD) and in transgenic mice that reproduce some features of this disease. In AD transgenic mice, alteration in olfaction has been associated with increased levels of soluble amyloid beta protein (Aβ) as well as with alterations in the oscillatory network activity recorded in the olfactory bulb (OB) and in the piriform cortex. However, since AD is a multifactorial disease and transgenic mice suffer a variety of adaptive changes, it is still unknown if soluble Aβ, by itself, is responsible for OB dysfunction both at electrophysiological and behavioral levels. Thus, here we tested whether or not Aβ directly affects OB network activity in vitro in slices obtained from mice and rats and if it affects olfactory ability in these rodents. Our results show that Aβ decreases, in a concentration- and time-dependent manner, the network activity of OB slices at clinically relevant concentrations (low nM) and in a reversible manner. Moreover, we found that intrabulbar injection of Aβ decreases the olfactory ability of rodents two weeks after application, an effect that is not related to alterations in motor performance or motivation to seek food and that correlates with the presence of Aβ deposits. Our results indicate that Aβ disrupts, at clinically relevant concentrations, the network activity of the OB in vitro and can trigger a disruption in olfaction. These findings open the possibility of exploring the cellular mechanisms involved in early pathological AD as an approach to reduce or halt its progress.
Collapse
Affiliation(s)
- Reynaldo Alvarado-Martínez
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, UNAM, Campus Juriquilla, Querétaro, México
| | - Karla Salgado-Puga
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, UNAM, Campus Juriquilla, Querétaro, México
| | - Fernando Peña-Ortega
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, UNAM, Campus Juriquilla, Querétaro, México
| |
Collapse
|
22
|
Kaushal A, Wani WY, Anand R, Gill KD. Spontaneous and induced nontransgenic animal models of AD: modeling AD using combinatorial approach. Am J Alzheimers Dis Other Demen 2013; 28:318-26. [PMID: 23687185 PMCID: PMC10852793 DOI: 10.1177/1533317513488914] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2024]
Abstract
Alzheimer's disease (AD), the most common neurodegenerative and dementing disorder, is characterized by extracellular amyloid deposition, intracellular neurofibrillary tangle formation, and neuronal loss. We are still behind in AD research in terms of knowledge regarding understanding its pathophysiology and designing therapeutics because of the lack of an accurate animal model for AD. A complete animal model of AD should imitate all the cognitive, behavioral, and neuropathological features of the disease. Partial models are currently in use, which only mimic specific and not all of the components of AD pathology. Currently the transgenic animals are the popular models for AD research, but different genetic backgrounds of these transgenic animals remain a major confounding factor. This review attempts to summarize the current literature on nontransgenic animal models of AD and to highlight the potential of exploiting spontaneous and induced animal models for neuropathological, neurochemical, neurobehavioral, and neuroprotective studies of AD.
Collapse
Affiliation(s)
- Alka Kaushal
- Department of Biochemistry,Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Willayat Yousuf Wani
- Department of Biochemistry,Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - R. Anand
- Department of Biochemistry,Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Kiran Dip Gill
- Department of Biochemistry,Post Graduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
23
|
Ihara Y, Morishima-Kawashima M, Nixon R. The ubiquitin-proteasome system and the autophagic-lysosomal system in Alzheimer disease. Cold Spring Harb Perspect Med 2012; 2:a006361. [PMID: 22908190 PMCID: PMC3405832 DOI: 10.1101/cshperspect.a006361] [Citation(s) in RCA: 130] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
As neurons age, their survival depends on eliminating a growing burden of damaged, potentially toxic proteins and organelles-a capability that declines owing to aging and disease factors. Here, we review the two proteolytic systems principally responsible for protein quality control in neurons and their important contributions to Alzheimer disease pathogenesis. In the first section, the discovery of paired helical filament ubiquitination is described as a backdrop for discussing the importance of the ubiquitin-proteasome system in Alzheimer disease. In the second section, we review the prominent involvement of the lysosomal system beginning with pathological endosomal-lysosomal activation and signaling at the very earliest stages of Alzheimer disease followed by the progressive failure of autophagy. These abnormalities, which result in part from Alzheimer-related genes acting directly on these lysosomal pathways, contribute to the development of each of the Alzheimer neuropathological hallmarks and represent a promising therapeutic target.
Collapse
Affiliation(s)
- Yasuo Ihara
- Department of Neuropathology, Faculty of Life and Medical Science, Doshisha University, Kyoto, Japan.
| | | | | |
Collapse
|
24
|
Han X, Ma Y, Liu X, Wang L, Qi S, Zhang Q, Du Y. Changes in insulin-signaling transduction pathway underlie learning/memory deficits in an Alzheimer’s disease rat model. J Neural Transm (Vienna) 2012; 119:1407-16. [DOI: 10.1007/s00702-012-0803-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2011] [Accepted: 03/29/2012] [Indexed: 12/20/2022]
|
25
|
Van Dam D, De Deyn PP. Animal models in the drug discovery pipeline for Alzheimer's disease. Br J Pharmacol 2012; 164:1285-300. [PMID: 21371009 DOI: 10.1111/j.1476-5381.2011.01299.x] [Citation(s) in RCA: 162] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
With increasing feasibility of predicting conversion of mild cognitive impairment to dementia based on biomarker profiling, the urgent need for efficacious disease-modifying compounds has become even more critical. Despite intensive research, underlying pathophysiological mechanisms remain insufficiently documented for purposeful target discovery. Translational research based on valid animal models may aid in alleviating some of the unmet needs in the current Alzheimer's disease pharmaceutical market, which includes disease-modification, increased efficacy and safety, reduction of the number of treatment unresponsive patients and patient compliance. The development and phenotyping of animal models is indeed essential in Alzheimer's disease-related research as valid models enable the appraisal of early pathological processes - which are often not accessible in patients, and subsequent target discovery and evaluation. This review paper summarizes and critically evaluates currently available animal models, and discusses their value to the Alzheimer drug discovery pipeline. Models dealt with include spontaneous models in various species, including senescence-accelerated mice, chemical and lesion-induced rodent models, and genetically modified models developed in Drosophila melanogaster, Caenorhabditis elegans, Danio rerio and rodents. Although highly valid animal models exist, none of the currently available models recapitulates all aspects of human Alzheimer's disease, and one should always be aware of the potential dangers of uncritical extrapolating from model organisms to a human condition that takes decades to develop and mainly involves higher cognitive functions.
Collapse
Affiliation(s)
- Debby Van Dam
- Laboratory of Neurochemistry & Behaviour, Institute Born-Bunge, Department of Biomedical Sciences, University of Antwerp, Wilrijk (Antwerp), Belgium.
| | | |
Collapse
|
26
|
Beck K, Schachtrup C. Vascular damage in the central nervous system: a multifaceted role for vascular-derived TGF-β. Cell Tissue Res 2011; 347:187-201. [PMID: 21850492 DOI: 10.1007/s00441-011-1228-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2011] [Accepted: 07/22/2011] [Indexed: 01/16/2023]
Abstract
The brain function depends on a continuous supply of blood. The blood-brain barrier (BBB), which is formed by vascular cells and glia, separates components of the circulating blood from neurons and maintains the precisely regulated brain milieu required for proper neuronal function. A compromised BBB alters the transport of molecules between the blood and brain and has been associated with or shown to precede neurodegenerative disease. Blood components immediately leak into the brain after mechanical damage or as a consequence of a compromised BBB in brain disease changing the extracellular environment at sites of vascular damage. It is intriguing how blood-derived components alter the cellular and molecular constituents of the neurovascular interface after BBB opening. We recently identified an unexpected role for the blood protein fibrinogen, which is deposited in the nervous system promptly after vascular damage, as an initial scar inducer by promoting the availability of active TGF-β. Fibrinogen-bound latent TGF-β interacts with astrocytes, leading to active TGF-β formation and activation of the TGF-β/Smad signaling pathway. Here, we discuss the pleiotropic effects of potentially vascular-derived TGF-β on cells at the neurovascular interface and we speculate how these biological effects might contribute to degeneration and regeneration processes. Summarizing the effects of the components derived from the brain vascular system on nervous system regeneration might support the development of new therapeutic approaches.
Collapse
Affiliation(s)
- Kristina Beck
- Centre of Chronic Immunodeficiency, University Medical Centre Freiburg and University of Freiburg, 79106 Freiburg, Germany
| | | |
Collapse
|
27
|
CD45 deficiency drives amyloid-β peptide oligomers and neuronal loss in Alzheimer's disease mice. J Neurosci 2011; 31:1355-65. [PMID: 21273420 DOI: 10.1523/jneurosci.3268-10.2011] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Converging lines of evidence indicate dysregulation of the key immunoregulatory molecule CD45 (also known as leukocyte common antigen) in Alzheimer's disease (AD). We report that transgenic mice overproducing amyloid-β peptide (Aβ) but deficient in CD45 (PSAPP/CD45(-/-) mice) faithfully recapitulate AD neuropathology. Specifically, we find increased abundance of cerebral intracellular and extracellular soluble oligomeric and insoluble Aβ, decreased plasma soluble Aβ, increased abundance of microglial neurotoxic cytokines tumor necrosis factor-α and interleukin-1β, and neuronal loss in PSAPP/CD45(-/-) mice compared with CD45-sufficient PSAPP littermates (bearing mutant human amyloid precursor protein and mutant human presenilin-1 transgenes). After CD45 ablation, in vitro and in vivo studies demonstrate an anti-Aβ phagocytic but proinflammatory microglial phenotype. This form of microglial activation occurs with elevated Aβ oligomers and neural injury and loss as determined by decreased ratio of anti-apoptotic Bcl-xL to proapoptotic Bax, increased activated caspase-3, mitochondrial dysfunction, and loss of cortical neurons in PSAPP/CD45(-/-) mice. These data show that deficiency in CD45 activity leads to brain accumulation of neurotoxic Aβ oligomers and validate CD45-mediated microglial clearance of oligomeric Aβ as a novel AD therapeutic target.
Collapse
|
28
|
|
29
|
Cheng YF, Wang C, Lin HB, Li YF, Huang Y, Xu JP, Zhang HT. Inhibition of phosphodiesterase-4 reverses memory deficits produced by Aβ25-35 or Aβ1-40 peptide in rats. Psychopharmacology (Berl) 2010; 212:181-91. [PMID: 20640406 DOI: 10.1007/s00213-010-1943-3] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2010] [Accepted: 06/25/2010] [Indexed: 01/28/2023]
Abstract
RATIONALE Cyclic AMP signaling plays an important role in memory loss associated with Alzheimer's disease (AD). However, little is known about whether inhibition of phosphodiesterase-4 (PDE4), which increases intracellular cAMP, reverses β-amyloid peptide (Aβ)-induced memory deficits. OBJECTIVE Experiments were performed to demonstrate the effect of the PDE4 inhibitor rolipram on memory impairment produced by Aβ1-40 (Aβ40) or its core fragment Aβ25-35. METHODS We tested memory using Morris water-maze and passive avoidance tasks and examined expression of phosphorylated cAMP response-element binding protein (pCREB) in the hippocampus in rats treated with Aβ25-35 or Aβ40 into bilateral CA1 subregions, with or without rolipram administration. RESULTS Aβ25-35 (10 μg/side) increased escape latency during acquisition training and decreased swimming time and distance in the target quadrant in the water-maze probe trial; it also decreased 24-h retention in the passive avoidance paradigm. All these were reversed by chronic administration of rolipram (0.5 mg/kg). Similarly, Aβ40 (4 μg/side) produced memory impairment, as demonstrated by decreased retention in passive avoidance; this was also reversed by repeated treatment with rolipram. In addition, rolipram blocked extinction of memory during the 32-day testing period in the passive avoidance test. Further, Aβ40 decreased pCREB expression in the hippocampus, which was also reversed by rolipram; the changes in pCREB were highly correlated with those in memory. CONCLUSIONS These results suggest that the PDE4 inhibitor rolipram reverses cognitive deficits associated with AD most likely via increased cAMP/CREB signaling in the hippocampus; PDE4 could be a target for drugs that improve cognition in AD.
Collapse
Affiliation(s)
- Yu-Fang Cheng
- Department of Pharmacology, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
30
|
Wang R, Malter JS, Wang DS. N-acetylcysteine prevents 4-hydroxynonenal- and amyloid-beta-induced modification and inactivation of neprilysin in SH-SY5Y cells. J Alzheimers Dis 2010; 19:179-89. [PMID: 20061637 DOI: 10.3233/jad-2010-1226] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
As one of the dominant amyloid-beta peptide (Abeta) proteases, neprilysin (NEP) plays a crucial role in maintaining a physiologic balance between Abeta production and catabolism. We have previously shown that NEP is modified by 4-hydroxynonenal (HNE) adducts, resulting in decreased activity in the brain of AD patients and cultured cells. In order to determine whether antioxidants can rescue NEP, SH-SY5Y cells were treated with HNE or Abeta, together with N-acetylcysteine for 24 hours, prior to analysis of NEP protein levels, activity, and oxidative modifications. Intracellular NEP developed HNE adducts after 24 hours of HNE or Abeta treatment as determined by immunoprecipitation, immunoblotting, and double immunofluorescence staining. N-acetylcysteine at 10 to 100 microM alleviated HNE adduction after HNE or Abeta treatment. In keeping with previous reports, HNE-modified NEP showed decreased catalytic activity. The present study demonstrates that antioxidants can be used to spare NEP from oxidative modification, suggesting a potential mechanism underlying the neuroprotective effects of antioxidants in aging or Alzheimer's disease.
Collapse
Affiliation(s)
- Rui Wang
- Department of Pathology and Laboratory Medicine, and Waisman Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706, USA
| | | | | |
Collapse
|
31
|
Cole GM, Frautschy SA. Mechanisms of action of non-steroidal anti-inflammatory drugs for the prevention of Alzheimer's disease. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2010; 9:140-8. [PMID: 20205646 DOI: 10.2174/187152710791011991] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2010] [Accepted: 02/22/2010] [Indexed: 01/05/2023]
Abstract
Alzheimer's disease (AD) is accompanied by an activation of the innate immune system, and many epidemiological studies have shown reduced risk for dementia or AD associated with chronic consumption of non-steroidal anti-inflammatory drugs (NSAIDs). These observations led to animal model studies to test the hypothesis that NSAIDs can be disease-modifying for some aspects of AD pathogenesis. NSAIDs cannot only suppress inflammatory targets, which could contribute to neuroprotection, they also slow amyloid deposition by mechanisms that remain unclear. Several large clinical trials with NSAID therapies with AD subjects have failed, and cyclooxygenase-2 does not appear to be a useful target for disease modifying therapy. However, there may be apolipoprotein E E4 pharmacogenomic effects and a real but delayed positive signal in a large primary prevention trial with naproxen. This encourages researchers to re-address possible mechanisms for a stage-dependent NSAID efficacy, the subject of this review.
Collapse
Affiliation(s)
- Greg M Cole
- Geriatric Research and Education Center, North Hills, CA 91343, USA
| | | |
Collapse
|
32
|
Kim EA, Kim H, Ahn JY, Hahn HG, Kim KS, Kim TU, Cho SW. Suppression of lipopolysaccharide-induced microglial activation by a benzothiazole derivative. Mol Cells 2010; 30:51-7. [PMID: 20652495 DOI: 10.1007/s10059-010-0087-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2010] [Revised: 03/11/2010] [Accepted: 03/25/2010] [Indexed: 12/28/2022] Open
Abstract
We previously reported that KHG21834, a benzothiazole derivative, attenuates the beta-amyloid (Abeta)-induced degeneration of both cortical and mesencephalic neurons in vitro. Central nervous system inflammation mediated by activated microglia is a key event in the development of neurodegenerative disease. In this study, we show that KHG21834 suppresses inflammation-mediated cytokine upregulation. Specifically, KHG21834 induces significant reductions in the lipopolysaccharide-induced activation of microglia and production of proinflammatory mediators such as tumor necrosis factor-alpha, interlukin-1beta, nitric oxide, and inducible nitric oxide synthase. In addition, KHG21834 blocks the expression of mitogen-activated protein kinases, including ERK, p38 MAPK, JNK, and Akt. In vivo intracerebroventricular infusion of KHG21834 also leads to decreases the level of interleukin-1beta and tumor necrosis factor-alpha in brain. These results, in combination with our previous findings on Abeta-induced degeneration, support the potential therapeutic efficacy of KHG21834 for the treatment of neurodegenerative disorders via the targeting of key glial activation pathways.
Collapse
Affiliation(s)
- Eun-A Kim
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 138-736, Korea
| | | | | | | | | | | | | |
Collapse
|
33
|
Kim EA, Hahn HG, Kim TU, Choi SY, Cho SW. Attenuation of β-amyloid-induced neuroinflammation by KHG21834 in vivo. BMB Rep 2010; 43:413-8. [DOI: 10.5483/bmbrep.2010.43.6.413] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
|
34
|
Cho S, Wood A, Bowlby MR. Brain slices as models for neurodegenerative disease and screening platforms to identify novel therapeutics. Curr Neuropharmacol 2010; 5:19-33. [PMID: 18615151 DOI: 10.2174/157015907780077105] [Citation(s) in RCA: 182] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2006] [Revised: 12/07/2006] [Accepted: 01/01/2007] [Indexed: 11/22/2022] Open
Abstract
Recent improvements in brain slice technology have made this biological preparation increasingly useful for examining pathophysiology of brain diseases in a tissue context. Brain slices maintain many aspects of in vivo biology, including functional local synaptic circuitry with preserved brain architecture, while allowing good experimental access and precise control of the extracellular environment, making them ideal platforms for dissection of molecular pathways underlying neuronal dysfunction. Importantly, these ex vivo systems permit direct treatment with pharmacological agents modulating these responses and thus provide surrogate therapeutic screening systems without recourse to whole animal studies. Virus or particle mediated transgenic expression can also be accomplished relatively easily to study the function of novel genes in a normal or injured brain tissue context.In this review we will discuss acute brain injury models in organotypic hippocampal and co-culture systems and the effects of pharmacological modulation on neurodegeneration. The review will also cover the evidence of developmental plasticity in these ex vivo models, demonstrating emergence of injury-stimulated neuronal progenitor cells, and neurite sprouting and axonal regeneration following pathway lesioning. Neuro-and axo-genesis are emerging as significant factors contributing to brain repair following many acute and chronic neurodegenerative disorders. Therefore brain slice models may provide a critical contextual experimental system to explore regenerative mechanisms in vitro.
Collapse
Affiliation(s)
- Seongeun Cho
- Discovery Neuroscience, Wyeth Research, CN8000, Princeton, NJ 08543, USA.
| | | | | |
Collapse
|
35
|
Sacrey LAR, Alaverdashvili M, Whishaw IQ. Similar hand shaping in reaching-for-food (skilled reaching) in rats and humans provides evidence of homology in release, collection, and manipulation movements. Behav Brain Res 2009; 204:153-61. [DOI: 10.1016/j.bbr.2009.05.035] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2009] [Revised: 05/26/2009] [Accepted: 05/27/2009] [Indexed: 11/16/2022]
|
36
|
|
37
|
Mitochondrial cholesterol loading exacerbates amyloid beta peptide-induced inflammation and neurotoxicity. J Neurosci 2009; 29:6394-405. [PMID: 19458211 DOI: 10.1523/jneurosci.4909-08.2009] [Citation(s) in RCA: 126] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The role of cholesterol in Alzheimer's disease (AD) has been linked to the generation of toxic amyloid beta peptides (Abeta). Using genetic mouse models of cholesterol loading, we examined whether mitochondrial cholesterol regulates Abeta neurotoxicity and AD pathology. Isolated mitochondria from brain or cortical neurons of transgenic mice overexpressing SREBP-2 (sterol regulatory element binding protein 2) or NPC1 (Niemann-Pick type C1) knock-out mice exhibited mitochondrial cholesterol accumulation, mitochondrial glutathione (mGSH) depletion and increased susceptibility to Abeta1-42-induced oxidative stress and release of apoptogenic proteins. Similar findings were observed in pharmacologically GSH-restricted rat brain mitochondria, while selective mGSH depletion sensitized human neuronal and glial cell lines to Abeta1-42-mediated cell death. Intracerebroventricular human Abeta delivery colocalized with mitochondria resulting in oxidative stress, neuroinflammation and neuronal damage that were enhanced in Tg-SREBP-2 mice and prevented upon mGSH recovery by GSH ethyl ester coinfusion, with a similar protection observed by intraperitoneal administration of GSH ethyl ester. Finally, APP/PS1 (amyloid precursor protein/presenilin 1) mice, a transgenic AD mouse model, exhibited mitochondrial cholesterol loading and mGSH depletion. Thus, mitochondrial cholesterol accumulation emerges as a novel pathogenic factor in AD by modulating Abeta toxicity via mGSH regulation; strategies boosting the particular pool of mGSH may be of relevance to slow down disease progression.
Collapse
|
38
|
Paul CM, Magda G, Abel S. Spatial memory: Theoretical basis and comparative review on experimental methods in rodents. Behav Brain Res 2009; 203:151-64. [PMID: 19467271 DOI: 10.1016/j.bbr.2009.05.022] [Citation(s) in RCA: 150] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2009] [Accepted: 05/17/2009] [Indexed: 02/08/2023]
Abstract
The assessment of learning and memory in animal models has been widely employed in scientific research for a long time. Among these models, those representing diseases with primary processes of affected memory - such as amnesia, dementia, brain aging, etc. - studies dealing with the toxic effects of specific drugs, and other exploring neurodevelopment, trauma, epilepsy and neuropsychiatric disorders, are often called on to employ these tools. There is a diversity of experimental methods assessing animal learning and memory skills. Overall, mazes are the devices mostly used today to test memory in rodents; there are several types of them, but their real usefulness, advantages and applications remain to be fully established and depend on the particular variant selected by the experimenter. The aims of the present article are first, to briefly review the accumulated knowledge in regard to spatial memory tasks; second, to bring the reader information on the different types of rodent mazes available to test spatial memory; and third, to elucidate the usefulness and limitations of each of these devices.
Collapse
Affiliation(s)
- Carrillo-Mora Paul
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City 14269, Mexico.
| | | | | |
Collapse
|
39
|
Wang R, Wang S, Malter JS, Wang DS. Effects of HNE-modification induced by Abeta on neprilysin expression and activity in SH-SY5Y cells. J Neurochem 2009; 108:1072-82. [PMID: 19196432 DOI: 10.1111/j.1471-4159.2008.05855.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The cerebral accumulation of beta-amyloid (Abeta) is a consistent feature of and likely contributor to the development of Alzheimer's disease. In addition to dysregulated production, increasing experimental evidence suggests reduced catabolism also plays an important role in Abeta accumulation. We have previously shown that neprilysin (NEP), the major protease which cleaves Abetain vivo, is modified by 4-hydroxy-nonenal (HNE) adducts in the brain of Alzheimer's disease patients. To determine if these changes affected Abeta, SH-SY5Y cells were treated with HNE or Abeta, and then NEP mRNA, protein levels, HNE adducted NEP, NEP activity and secreted Abeta levels were determined. Intracellular NEP developed HNE adducts after 24 h of HNE treatment as determined by immunoprecipitation, immunoblotting, and double immunofluorescence staining. HNE-modified NEP showed decreased catalytic activity, which was associated with elevations in Abeta1-40 in SH-SY5Y and H4 APP695wt cells. Incubation of cells with Abeta1-42 also induced HNE adduction of NEP. In an apparent compensatory response, Abeta-treated cells showed increased NEP mRNA and protein expression. Despite elevations in NEP protein, the activity was significantly lower compared with the NEP protein level. This study demonstrates that NEP can be inactivated by HNE-adduction, which is associated with, at least partly, reduced Abeta cleavage and enhanced Abeta accumulation.
Collapse
Affiliation(s)
- Rui Wang
- Department of Pathology, School of Medicine and Public Health, University of Wisconsin, Madison, USA
| | | | | | | |
Collapse
|
40
|
Catania C, Sotiropoulos I, Silva R, Onofri C, Breen KC, Sousa N, Almeida OFX. The amyloidogenic potential and behavioral correlates of stress. Mol Psychiatry 2009; 14:95-105. [PMID: 17912249 DOI: 10.1038/sj.mp.4002101] [Citation(s) in RCA: 134] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Observations of elevated basal cortisol levels in Alzheimer's disease (AD) patients prompted the hypothesis that stress and glucocorticoids (GC) may contribute to the development and/or maintenance of AD. Consistent with that hypothesis, we show that stress and GC provoke misprocessing of amyloid precursor peptide in the rat hippocampus and prefrontal cortex, resulting in increased levels of the peptide C-terminal fragment 99 (C99), whose further proteolytic cleavage results in the generation of amyloid-beta (Abeta). We also show that exogenous Abeta can reproduce the effects of stress and GC on C99 production and that a history of stress strikingly potentiates the C99-inducing effects of Abeta and GC. Previous work has indicated a role for Abeta in disruption of synaptic function and cognitive behaviors, and AD patients reportedly show signs of heightened anxiety. Here, behavioral analysis revealed that like stress and GC, Abeta administration causes spatial memory deficits that are exacerbated by stress and GC; additionally, Abeta, stress and GC induced a state of hyperanxiety. Given that the intrinsic properties of C99 and Abeta include neuroendangerment and behavioral impairment, our findings suggest a causal role for stress and GC in the etiopathogenesis of AD, and demonstrate that stressful life events and GC therapy can have a cumulative impact on the course of AD development and progression.
Collapse
Affiliation(s)
- C Catania
- Max Planck Institute of Psychiatry, Munich, Germany
| | | | | | | | | | | | | |
Collapse
|
41
|
Begum AN, Yang F, Teng E, Hu S, Jones MR, Rosario ER, Beech W, Hudspeth B, Ubeda OJ, Cole GM, Frautschy SA. Use of copper and insulin-resistance to accelerate cognitive deficits and synaptic protein loss in a rat Abeta-infusion Alzheimer's disease model. J Alzheimers Dis 2008; 15:625-40. [PMID: 19096161 PMCID: PMC4313743 DOI: 10.3233/jad-2008-15409] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The rat amyloid-beta (Abeta) intracerebroventricular infusion can model aspects of Alzheimer's disease (AD) and has predicted efficacy of therapies such as ibuprofen and curcumin in transgenic mouse models. High density lipoprotein (HDL), a normal plasma carrier of Abeta, is used to attenuate Abeta aggregation within the pump, causing Abeta-dependent toxicity and cognitive deficits within 3 months. Our goal was to identify factors that might accelerate onset of Abeta-dependent deficits to improve efficiency and cost-effectiveness of model. We focused on: 1) optimizing HDL-Abeta preparation for maximal toxicity; 2) evaluating the role of copper, a factor typically in water that can impact oligomer stability; and 3) determining impact of insulin resistance (type II diabetes), a risk factor for AD. In vitro studies were performed to determine doses of copper and methods of Abeta-HDL preparation that maximized toxicity. These preparations when infused resulted in earlier onset of cognitive deficits within 6 weeks post-infusion. Induction of insulin resistance did not exacerbate Abeta-dependent cognitive deficits, but did exacerbate synaptic protein loss. In summary, the newly described in vivo infusion model may be useful cost-effective method for screening for new therapeutic drugs for AD.
Collapse
Affiliation(s)
- Aynun N. Begum
- Department of Medicine, University of California, Los Angeles, CA 90095, USA
- Geriatric Research Education Clinical Center (GRECC), North Hills, CA, USA
| | - Fusheng Yang
- Department of Medicine, University of California, Los Angeles, CA 90095, USA
- Geriatric Research Education Clinical Center (GRECC), North Hills, CA, USA
| | - Edmond Teng
- Department of Neurology, University of California, Los Angeles, CA 90095, USA
- Greater Los Angeles Veterans Affairs Healthcare System, Neurobehavior Unit, Los Angeles, CA, USA
| | - Shuxin Hu
- Department of Medicine, University of California, Los Angeles, CA 90095, USA
- Geriatric Research Education Clinical Center (GRECC), North Hills, CA, USA
| | - Mychica R. Jones
- Department of Medicine, University of California, Los Angeles, CA 90095, USA
- Geriatric Research Education Clinical Center (GRECC), North Hills, CA, USA
| | - Emily R. Rosario
- Department of Medicine, University of California, Los Angeles, CA 90095, USA
- Geriatric Research Education Clinical Center (GRECC), North Hills, CA, USA
| | - Walter Beech
- Department of Medicine, University of California, Los Angeles, CA 90095, USA
- Geriatric Research Education Clinical Center (GRECC), North Hills, CA, USA
| | - Beverly Hudspeth
- Department of Medicine, University of California, Los Angeles, CA 90095, USA
- Geriatric Research Education Clinical Center (GRECC), North Hills, CA, USA
| | - Oliver J. Ubeda
- Department of Medicine, University of California, Los Angeles, CA 90095, USA
- Geriatric Research Education Clinical Center (GRECC), North Hills, CA, USA
| | - Greg M. Cole
- Department of Medicine, University of California, Los Angeles, CA 90095, USA
- Department of Neurology, University of California, Los Angeles, CA 90095, USA
- Geriatric Research Education Clinical Center (GRECC), North Hills, CA, USA
| | - Sally A. Frautschy
- Department of Medicine, University of California, Los Angeles, CA 90095, USA
- Department of Neurology, University of California, Los Angeles, CA 90095, USA
- Geriatric Research Education Clinical Center (GRECC), North Hills, CA, USA
| |
Collapse
|
42
|
Zaheer A, Zaheer S, Thangavel R, Wu Y, Sahu SK, Yang B. Glia maturation factor modulates beta-amyloid-induced glial activation, inflammatory cytokine/chemokine production and neuronal damage. Brain Res 2008; 1208:192-203. [PMID: 18395194 DOI: 10.1016/j.brainres.2008.02.093] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2008] [Revised: 02/19/2008] [Accepted: 02/24/2008] [Indexed: 01/01/2023]
Abstract
Glia maturation factor (GMF), discovered and characterized in our laboratory, is a highly conserved protein primarily localized in mammalian central nervous system. Previously we demonstrated that GMF is required in the induced production of proinflammatory cytokines and chemokines in brain cells. We now report that ventricular infusion of human amyloid beta peptide1-42 (Abeta1-42) in mouse brain caused glial activation and large increases in the levels of GMF as well as induction of inflammatory cytokine/chemokine known for launching the neuro inflammatory cascade in Alzheimer's disease (AD). To test the hypothesis that GMF is involved in the pathogenesis of AD, we infused Abeta1-42 in the brain of GMF-deficient (GMF-KO) mice, recently prepared in our laboratory. GMF-deficient mice showed reduced glial activation and significantly suppressed proinflammatory cytokine/chemokine production following Abeta infusion compared to wild type (Wt) mice. The decrease in glial activation in the GMF-KO mice is also associated with significant reduction in Abeta induced loss of pre-synaptic marker, synaptophysin, and post-synaptic density protein-95 (PSD 95). We also examined the potential relationship between GMF or lack of it with learning and memory using the T-maze, Y-maze, and water maze, hippocampal-dependent spatial memory tasks. Our results show that memory retention was improved in GMF-KO mice compared to Wt controls following Abeta infusion. Diminution of these Abeta1-42 effects in primary cultures of GMF-KO astrocyte and microglia were reversed by reconstituted expression of GMF. Taken together, our results indicate a novel mediatory role of GMF in the neuro-inflammatory pathway of Abeta and its pro-inflammatory functions.
Collapse
Affiliation(s)
- Asgar Zaheer
- Veterans Affair Medical Center, and Division of Neurochemistry and Neurobiology, Department of Neurology, University of Iowa, Iowa City, IA 52242, USA.
| | | | | | | | | | | |
Collapse
|
43
|
Bonin M, Marx FP, Kautzmann S, Riess O, Krüger R. Microarray expression analysis reveals genetic pathways implicated in C621 synphilin-1-mediated toxicity. J Neural Transm (Vienna) 2008; 115:941-58. [DOI: 10.1007/s00702-008-0031-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2007] [Accepted: 02/01/2008] [Indexed: 10/22/2022]
|
44
|
Du D, Chen S, Cai J, Song D. Comparison of drug sensitivity using acetylcholinesterase biosensor based on nanoparticles–chitosan sol–gel composite. J Electroanal Chem (Lausanne) 2007. [DOI: 10.1016/j.jelechem.2007.08.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
45
|
Iribarren P, Chen K, Gong W, Cho EH, Lockett S, Uranchimeg B, Wang JM. Interleukin 10 and TNFalpha synergistically enhance the expression of the G protein-coupled formylpeptide receptor 2 in microglia. Neurobiol Dis 2007; 27:90-8. [PMID: 17544285 PMCID: PMC1989777 DOI: 10.1016/j.nbd.2007.04.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2006] [Revised: 04/11/2007] [Accepted: 04/27/2007] [Indexed: 11/28/2022] Open
Abstract
Microglia are important participants in inflammatory responses in the central nervous system. We previously observed that tumor necrosis factor alpha (TNFalpha) induces the expression of the formylpeptide receptor mFPR2 on microglial cells. This chemoattractant receptor mediates microglial cell chemotaxis in response to a variety of peptides, including amyloid beta peptide (Abeta(42)), a major pathogenic factor in Alzheimer's disease (AD). In search for agents that regulate microglial activation, we unexpectedly found that IL-10 enhanced the expression of mFPR2 on TNFalpha-activated microglia. This was associated with a markedly increased microglial chemotaxis to Abeta(42) and its endocytosis via mFPR2. Mechanistic studies revealed that the synergistic effect of IL-10 on TNFalpha-induction of mFPR2 in microglia was dependent on activation of p38 MAPK. Our results suggest that IL-10 may affect the pathogenic process of AD by up-regulating mFPR2 and thus favoring the recognition and internalization of Abeta(42) by activated microglial cells.
Collapse
Affiliation(s)
- Pablo Iribarren
- Laboratory of Molecular Immunoregulation, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, USA.
| | | | | | | | | | | | | |
Collapse
|
46
|
Craft JM, Watterson DM, Van Eldik LJ. Human amyloid β-induced neuroinflammation is an early event in neurodegeneration. Glia 2006; 53:484-90. [PMID: 16369931 DOI: 10.1002/glia.20306] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Using a human amyloid beta (Abeta) intracerebroventricular infusion mouse model of Alzheimer's disease-related injury, we previously demonstrated that systemic administration of a glial activation inhibitor could suppress neuroinflammation, prevent synaptic damage, and attenuate hippocampal-dependent behavioral deficits. We report that Abeta-induced neuroinflammation is an early event associated with onset and progression of pathophysiology, can be suppressed by the glial inhibitor over a range of intervention start times, and is amenable to suppression without inhibiting peripheral tissue inflammatory responses. Specifically, hippocampal neuroinflammation and neurodegeneration occur in close time proximity at 4-6 weeks after the start of infusion. Intraperitoneal administration of inhibitor for 2-week intervals starting at various times after initiation of Abeta infusion suppresses progression of pathophysiology. The glial inhibitor is a selective suppressor of neuroinflammation, in that it does not block peripheral tissue production of proinflammatory cytokines or markers of B- and T-cell activation after a systemic lipopolysaccharide challenge. These results support a causal link between neuroinflammation and neurodegeneration, have important implications for future therapeutic development, and provide insight into the relative time window for targeting neuroinflammation with positive neurological outcomes.
Collapse
Affiliation(s)
- Jeffrey M Craft
- Center for Drug Discovery and Chemical Biology, Northwestern University, Chicago, Illinois 60611, USA
| | | | | |
Collapse
|
47
|
Le Y, Yu X, Ruan L, Wang O, Qi D, Zhu J, Lu X, Kong Y, Cai K, Pang S, Shi X, Wang JM. The immunopharmacological properties of transforming growth factor beta. Int Immunopharmacol 2005; 5:1771-82. [PMID: 16275614 DOI: 10.1016/j.intimp.2005.07.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2005] [Revised: 06/28/2005] [Accepted: 07/18/2005] [Indexed: 02/02/2023]
Abstract
Transforming growth factor-beta (TGF-beta) family members are multifunctional molecules, which play pivotal roles in regulating cell proliferation, differentiation, migration, development, tissue remodeling and repair. These events are closely associated with host immune responses and inflammation. Despite some controversies on their function in controlling dendritic and T regulatory cell development and activity, the importance of TGF-betas in the progress of autoimmunity and inflammatory diseases has been well appreciated and new aspects of their contribution continue to be recognized. Since one of the major biological properties of TGF-betas is its capacity to potently suppress immune responses, they are considered as candidates for the development of therapeutic agents to fend off undesirable damage associated with immune and inflammatory conditions.
Collapse
Affiliation(s)
- Yingying Le
- Laboratory of Immunologic and Inflammatory Diseases, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, P.R. China.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Xie Z, Harris-White ME, Wals PA, Frautschy SA, Finch CE, Morgan TE. Apolipoprotein J (clusterin) activates rodent microglia in vivo and in vitro. J Neurochem 2005; 93:1038-46. [PMID: 15857407 DOI: 10.1111/j.1471-4159.2005.03065.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Apolipoprotein J (apoJ; also known as clusterin and sulfated glycoprotein (SGP)-2) is associated with senile plaques in degenerating regions of Alzheimer's disease brains, where activated microglia are also prominent. We show a functional link between apoJ and activated microglia by demonstrating that exogenous apoJ activates rodent microglia in vivo and in vitro. Intracerebroventricular infusion of purified human plasma apoJ ( approximately 4 microg over 28 days) activated parenchymal microglia to a phenotype characterized by enlarged cell bodies and processes (phosphotyrosine immunostaining). In vitro, primary rat microglia were also activated by apoJ, with changes in morphology and induction of major histocompatibility complex class II (MHCII) antigen. ApoJ increased the secretion of reactive nitrogen intermediates in a dose-dependent manner (EC(50) 112 nm), which was completely blocked by aminoguanidine (AG), a nitric oxide synthase inhibitor. However, AG did not block the increased secretion of tumor necrosis factor-alpha by apoJ (EC(50) 55 nm). Microglial activation by apoJ was also blocked by an anti-apoJ monoclonal antibody (G7), and by chemical cleavage of apoJ with 2-nitro-5-thiocyanobenzoate. The mitogen-activated protein kinase kinase and protein kinase C inhibitors PD98059 and H7 inhibited apoJ-mediated induction of reactive nitrogen intermediate secretion from cultured microglia. As a functional measure, apoJ-activated microglia secreted neurotoxic agents in a microglia-neuron co-culture model. We hypothesize that ApoJ contributes to chronic inflammation and neurotoxicity through direct effects on microglia.
Collapse
Affiliation(s)
- Z Xie
- Andrus Gerontology Center and Department of Biological Sciences, University of Southern California, Los Angeles, 90089, USA
| | | | | | | | | | | |
Collapse
|
49
|
Craft JM, Watterson DM, Hirsch E, Van Eldik LJ. Interleukin 1 receptor antagonist knockout mice show enhanced microglial activation and neuronal damage induced by intracerebroventricular infusion of human beta-amyloid. J Neuroinflammation 2005; 2:15. [PMID: 15967035 PMCID: PMC1190207 DOI: 10.1186/1742-2094-2-15] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2005] [Accepted: 06/20/2005] [Indexed: 12/14/2022] Open
Abstract
Background Interleukin 1 (IL-1) is a key mediator of immune responses in health and disease. Although classically the function of IL-1 has been studied in the systemic immune system, research in the past decade has revealed analogous roles in the CNS where the cytokine can contribute to the neuroinflammation and neuropathology seen in a number of neurodegenerative diseases. In Alzheimer's disease (AD), for example, pre-clinical and clinical studies have implicated IL-1 in the progression of a pathologic, glia-mediated pro-inflammatory state in the CNS. The glia-driven neuroinflammation can lead to neuronal damage, which, in turn, stimulates further glia activation, potentially propagating a detrimental cycle that contributes to progression of pathology. A prediction of this neuroinflammation hypothesis is that increased IL-1 signaling in vivo would correlate with increased severity of AD-relevant neuroinflammation and neuronal damage. Methods To test the hypothesis that increased IL-1 signaling predisposes animals to beta-amyloid (Aβ)-induced damage, we used IL-1 receptor antagonist Knock-Out (IL1raKO) and wild-type (WT) littermate mice in a model that involves intracerebroventricular infusion of human oligomeric Aβ1–42. This model mimics many features of AD, including robust neuroinflammation, Aβ plaques, synaptic damage and neuronal loss in the hippocampus. IL1raKO and WT mice were infused with Aβ for 28 days, sacrificed at 42 days, and hippocampal endpoints analyzed. Results IL1raKO mice showed increased vulnerability to Aβ-induced neuropathology relative to their WT counterparts. Specifically, IL1raKO mice exhibited increased mortality, enhanced microglial activation and neuroinflammation, and more pronounced loss of synaptic markers. Interestingly, Aβ-induced astrocyte responses were not significantly different between WT and IL1raKO mice, suggesting that enhanced IL-1 signaling predominately affects microglia. Conclusion Our data are consistent with the neuroinflammation hypothesis whereby increased IL-1 signaling in AD enhances glia activation and leads to an augmented neuroinflammatory process that increases the severity of neuropathologic sequelae.
Collapse
Affiliation(s)
- Jeffrey M Craft
- Center for Drug Discovery and Chemical Biology, Northwestern University, Chicago, IL, USA
- Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - D Martin Watterson
- Center for Drug Discovery and Chemical Biology, Northwestern University, Chicago, IL, USA
- Molecular Pharmacology and Biological Chemistry, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Emmet Hirsch
- Obstetrics and Gynecology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Obstetrics and Gynecology, Evanston Northwestern Healthcare, Evanston, IL, USA
| | - Linda J Van Eldik
- Center for Drug Discovery and Chemical Biology, Northwestern University, Chicago, IL, USA
- Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
50
|
Hamaguchi T, Okino S, Sodeyama N, Itoh Y, Takahashi A, Otomo E, Matsushita M, Mizusawa H, Yamada M. Association of a polymorphism of the transforming growth factor-beta1 gene with cerebral amyloid angiopathy. J Neurol Neurosurg Psychiatry 2005; 76:696-9. [PMID: 15834029 PMCID: PMC1739647 DOI: 10.1136/jnnp.2003.034454] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
BACKGROUND A recent study showed that transforming growth factor-beta1 (TGF-beta1) induces amyloid-beta deposition in cerebral blood vessels and meninges of a transgenic mouse model of Alzheimer's disease (AD), and that TGF-beta1 mRNA levels are correlated with cerebral amyloid angiopathy (CAA) in human AD brains. A T/C polymorphism at codon 10 in exon 1 of the TGF-beta1 gene has been reported to be associated with the serum TGF-beta1 concentration. We investigated whether the TGF-beta1 polymorphism is associated with the risk of CAA. METHODS The association between the severity of CAA and the T/C polymorphism at codon 10 in exon 1 of the TGF-beta1 was investigated in 167 elderly Japanese autopsy cases, including 73 patients with AD. The apolipoprotein E (APOE) genotype was also determined. RESULTS The genotypes (TT/ TC/ CC) were associated with the severity of CAA significantly in all patients (p = 0.0026), in non-AD patients (p = 0.011), and APOE non-epsilon4 carriers (p = 0.0099), but not in AD patients or APOE epsilon4 carriers. The number of the T alleles positively correlated with the severity of CAA in all patients (p = 0.0011), non-AD patients (p = 0.0026), and APOE non-epsilon4 carriers (p = 0.0028), but not in AD patients or APOE epsilon4 carriers. The polymorphism was not significantly associated with AD. CONCLUSIONS Our results suggest that the polymorphism in TGF-beta1 is associated with the severity of CAA, especially in non-AD patients and APOE non-epsilon4 carriers.
Collapse
Affiliation(s)
- T Hamaguchi
- Department of Neurology and Neurobiology of Aging, Kanazawa University Graduate School of Medical Science, 13-1 Takara-machi, Kanazawa 920-8640, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|