1
|
Iwata N, Tsubuki S, Sekiguchi M, Watanabe-Iwata K, Matsuba Y, Kamano N, Fujioka R, Takamura R, Watamura N, Kakiya N, Mihira N, Morito T, Shirotani K, Mann DM, Robinson AC, Hashimoto S, Sasaguri H, Saito T, Higuchi M, Saido TC. Metabolic resistance of Aβ3pE-42, a target epitope of the anti-Alzheimer therapeutic antibody, donanemab. Life Sci Alliance 2024; 7:e202402650. [PMID: 39348937 PMCID: PMC11443169 DOI: 10.26508/lsa.202402650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 09/06/2024] [Accepted: 09/06/2024] [Indexed: 10/02/2024] Open
Abstract
The amyloid β peptide (Aβ), starting with pyroglutamate (pE) at position 3 and ending at position 42 (Aβ3pE-42), predominantly accumulates in the brains of Alzheimer's disease. Consistently, donanemab, a therapeutic antibody raised against Aβ3pE-42, has been shown to be effective in recent clinical trials. Although the primary Aβ produced physiologically is Aβ1-40/42, an explanation for how and why this physiological Aβ is converted to the pathological form remains elusive. Here, we present experimental evidence that accounts for the aging-associated Aβ3pE-42 deposition: Aβ3pE-42 was metabolically more stable than other Aβx-42 variants; deficiency of neprilysin, the major Aβ-degrading enzyme, induced a relatively selective deposition of Aβ3pE-42 in both APP transgenic and App knock-in mouse brains; Aβ3pE-42 deposition always colocalized with Pittsburgh compound B-positive cored plaques in APP transgenic mouse brains; and under aberrant conditions, such as a significant reduction in neprilysin activity, aminopeptidases, dipeptidyl peptidases, and glutaminyl-peptide cyclotransferase-like were up-regulated in the progression of aging, and a proportion of Aβ1-42 may be processed to Aβ3pE-42. Our findings suggest that anti-Aβ therapies are more effective if given before Aβ3pE-42 deposition.
Collapse
Affiliation(s)
- Nobuhisa Iwata
- Department of Genome-Based Drug Discovery and Leading Medical Research Core Unit, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama, Japan
| | - Satoshi Tsubuki
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama, Japan
| | - Misaki Sekiguchi
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama, Japan
| | - Kaori Watanabe-Iwata
- Department of Genome-Based Drug Discovery and Leading Medical Research Core Unit, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Yukio Matsuba
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama, Japan
| | - Naoko Kamano
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama, Japan
| | - Ryo Fujioka
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama, Japan
| | - Risa Takamura
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama, Japan
| | - Naoto Watamura
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama, Japan
| | - Naomasa Kakiya
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama, Japan
| | - Naomi Mihira
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama, Japan
| | - Takahiro Morito
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama, Japan
| | - Keiro Shirotani
- Department of Genome-Based Drug Discovery and Leading Medical Research Core Unit, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - David Ma Mann
- Division of Neuroscience, Faculty of Biology, Medicine and Health, School of Biological Sciences, Faculty of Biology, Medicine and Health, School of Biological Sciences, The University of Manchester, Salford Royal Hospital, Salford, UK
| | - Andrew C Robinson
- Division of Neuroscience, Faculty of Biology, Medicine and Health, School of Biological Sciences, Faculty of Biology, Medicine and Health, School of Biological Sciences, The University of Manchester, Salford Royal Hospital, Salford, UK
| | - Shoko Hashimoto
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama, Japan
| | - Hiroki Sasaguri
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama, Japan
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Makoto Higuchi
- Department of Functional Brain Imaging, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama, Japan
| |
Collapse
|
2
|
Karaahmet B, Zhang Y, Duquesne L, Sigalov A, Yung C, Kroshilina A, Bennett DA, Taga M, Klein HU. Spatial Transcriptomic Analysis Identifies a SERPINA3-Expressing Astrocytic State Associated with the Human Neuritic Plaque Microenvironment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.13.623438. [PMID: 39605680 PMCID: PMC11601401 DOI: 10.1101/2024.11.13.623438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Single-nucleus transcriptomic studies have revealed glial cell states associated with Alzheimer's disease; however, these nuclei are dissociated from the complex architecture of the human neocortex. Here, we successfully performed an unbiased distance-based analytic strategy on spatially-registered transcriptomic data. Leveraging immunohistochemistry in the same tissue section, our analyses prioritized SERPINA3 and other genes, such as metallothioneins, as altered in the vicinity of neuritic amyloid plaques. Results were validated at the protein level by immunofluorescence, highlighting that a reactive SERPINA3+ astrocyte subtype, Ast.5, plays a role in the plaque microenvironment.
Collapse
Affiliation(s)
- Berke Karaahmet
- Center for Translational & Computational Neuroimmunology, Department of Neurology and Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Medical Center, New York, New York, USA
| | - Ya Zhang
- Center for Translational & Computational Neuroimmunology, Department of Neurology and Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Medical Center, New York, New York, USA
| | - Laurine Duquesne
- Center for Translational & Computational Neuroimmunology, Department of Neurology and Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Medical Center, New York, New York, USA
| | - Alina Sigalov
- Center for Translational & Computational Neuroimmunology, Department of Neurology and Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Medical Center, New York, New York, USA
| | - Christina Yung
- Center for Translational & Computational Neuroimmunology, Department of Neurology and Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Medical Center, New York, New York, USA
| | - Alexandra Kroshilina
- Center for Translational & Computational Neuroimmunology, Department of Neurology and Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Medical Center, New York, New York, USA
| | - David A. Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Mariko Taga
- Center for Translational & Computational Neuroimmunology, Department of Neurology and Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Medical Center, New York, New York, USA
| | - Hans-Ulrich Klein
- Center for Translational & Computational Neuroimmunology, Department of Neurology and Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Medical Center, New York, New York, USA
| |
Collapse
|
3
|
Ma K, Zhang C, Zhang H, An C, Li G, Cheng L, Li M, Ren M, Bai Y, Liu Z, Ji S, Liu X, Gao J, Zhang Z, Wu X, Chen X. High-Salt Diet Accelerates Neuron Loss and Anxiety in APP/PS1 Mice Through Serpina3n. Int J Mol Sci 2024; 25:11731. [PMID: 39519278 PMCID: PMC11546851 DOI: 10.3390/ijms252111731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/29/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
High salt (HS) consumption is an independent risk factor for neurodegenerative diseases such as dementia, stroke, and cerebral small vessel disease related to cognitive decline. Recently, Alzheimer's disease-like pathology changes have been reported as consequences of a HS diet in wild-type (wt) mice. However, it has not been revealed how HS diets accelerate the progress of Alzheimer's disease (AD) in APP/PS1 mice. Here, we fed APP/PS1 mice a HS diet or normal diet (ND) for six months; the effects of the HS/ND on wt mice were also observed. The results of our behavior test reveal that the HS diet exacerbates anxiety, β-amyloid overload, neuron loss, and synapse damage in the hippocampi of APP/PS1 mice; this was not observed in HS-treated wt mice. RNA sequencing shows that nearly all serpin family members were increased in the hippocampus of HS-treated APP/PS1 mice. Gene function analysis showed that a HS diet induces neurodegeneration, including axon dysfunction and neuro-ligand-based dysfunction, and regulates serine protein inhibitor activities. The mRNA and protein levels of Serpina3n were dramatically increased. Upregulated Serpina3n may be the key for β-amyloid aggregation and neuronal loss in the hippocampus of HS-treated APP/PS1 mice. Serpina3n inhibition attenuated the anxiety and increased the number of neurons in the hippocampal CA1(cornu ammonis) region of APP/PS1 mice. Our study provides novel insights into the mechanisms by which excessive HS diet deteriorates anxiety in AD mice. Therefore, decreasing daily dietary salt consumption constitutes a pivotal public health intervention for mitigating the progression of neuropathology, especially for old patients and those with neurodegenerative disease.
Collapse
Affiliation(s)
- Kaige Ma
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China; (K.M.); (C.Z.); (H.Z.); (C.A.); (G.L.); (L.C.); (M.L.); (M.R.); (Y.B.); (Z.L.); (S.J.); (X.L.); (J.G.); (Z.Z.)
- Key Laboratory of Ministry of Education for Environment and Genes Related to Diseases, Xi’an Jiaotong University, Xi’an 710061, China
| | - Chenglin Zhang
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China; (K.M.); (C.Z.); (H.Z.); (C.A.); (G.L.); (L.C.); (M.L.); (M.R.); (Y.B.); (Z.L.); (S.J.); (X.L.); (J.G.); (Z.Z.)
| | - Hanyue Zhang
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China; (K.M.); (C.Z.); (H.Z.); (C.A.); (G.L.); (L.C.); (M.L.); (M.R.); (Y.B.); (Z.L.); (S.J.); (X.L.); (J.G.); (Z.Z.)
- Key Laboratory of Ministry of Education for Environment and Genes Related to Diseases, Xi’an Jiaotong University, Xi’an 710061, China
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China
| | - Chanyuan An
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China; (K.M.); (C.Z.); (H.Z.); (C.A.); (G.L.); (L.C.); (M.L.); (M.R.); (Y.B.); (Z.L.); (S.J.); (X.L.); (J.G.); (Z.Z.)
| | - Ge Li
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China; (K.M.); (C.Z.); (H.Z.); (C.A.); (G.L.); (L.C.); (M.L.); (M.R.); (Y.B.); (Z.L.); (S.J.); (X.L.); (J.G.); (Z.Z.)
| | - Lixue Cheng
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China; (K.M.); (C.Z.); (H.Z.); (C.A.); (G.L.); (L.C.); (M.L.); (M.R.); (Y.B.); (Z.L.); (S.J.); (X.L.); (J.G.); (Z.Z.)
- Key Laboratory of Ministry of Education for Environment and Genes Related to Diseases, Xi’an Jiaotong University, Xi’an 710061, China
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China
| | - Mai Li
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China; (K.M.); (C.Z.); (H.Z.); (C.A.); (G.L.); (L.C.); (M.L.); (M.R.); (Y.B.); (Z.L.); (S.J.); (X.L.); (J.G.); (Z.Z.)
| | - Minghe Ren
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China; (K.M.); (C.Z.); (H.Z.); (C.A.); (G.L.); (L.C.); (M.L.); (M.R.); (Y.B.); (Z.L.); (S.J.); (X.L.); (J.G.); (Z.Z.)
| | - Yudan Bai
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China; (K.M.); (C.Z.); (H.Z.); (C.A.); (G.L.); (L.C.); (M.L.); (M.R.); (Y.B.); (Z.L.); (S.J.); (X.L.); (J.G.); (Z.Z.)
- Key Laboratory of Ministry of Education for Environment and Genes Related to Diseases, Xi’an Jiaotong University, Xi’an 710061, China
| | - Zichang Liu
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China; (K.M.); (C.Z.); (H.Z.); (C.A.); (G.L.); (L.C.); (M.L.); (M.R.); (Y.B.); (Z.L.); (S.J.); (X.L.); (J.G.); (Z.Z.)
| | - Shengfeng Ji
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China; (K.M.); (C.Z.); (H.Z.); (C.A.); (G.L.); (L.C.); (M.L.); (M.R.); (Y.B.); (Z.L.); (S.J.); (X.L.); (J.G.); (Z.Z.)
| | - Xiyue Liu
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China; (K.M.); (C.Z.); (H.Z.); (C.A.); (G.L.); (L.C.); (M.L.); (M.R.); (Y.B.); (Z.L.); (S.J.); (X.L.); (J.G.); (Z.Z.)
| | - Jinman Gao
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China; (K.M.); (C.Z.); (H.Z.); (C.A.); (G.L.); (L.C.); (M.L.); (M.R.); (Y.B.); (Z.L.); (S.J.); (X.L.); (J.G.); (Z.Z.)
| | - Zhichao Zhang
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China; (K.M.); (C.Z.); (H.Z.); (C.A.); (G.L.); (L.C.); (M.L.); (M.R.); (Y.B.); (Z.L.); (S.J.); (X.L.); (J.G.); (Z.Z.)
| | - Xiaolin Wu
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China
- Institute of Neuroscience, Translational Medicine Institute, Xi’an Jiaotong University, Xi’an 710061, China
| | - Xinlin Chen
- Department/Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an 710061, China; (K.M.); (C.Z.); (H.Z.); (C.A.); (G.L.); (L.C.); (M.L.); (M.R.); (Y.B.); (Z.L.); (S.J.); (X.L.); (J.G.); (Z.Z.)
- Key Laboratory of Ministry of Education for Environment and Genes Related to Diseases, Xi’an Jiaotong University, Xi’an 710061, China
| |
Collapse
|
4
|
Reyes CDG, Mojgan A, Fowowe M, Onigbinde S, Daramola O, Lubman DM, Mechref Y. Differential expression of N-glycopeptides derived from serum glycoproteins in mild cognitive impairment (MCI) patients. Proteomics 2024; 24:e2300620. [PMID: 38602241 PMCID: PMC11749004 DOI: 10.1002/pmic.202300620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 03/15/2024] [Accepted: 03/19/2024] [Indexed: 04/12/2024]
Abstract
Mild cognitive impairment (MCI) is an early stage of memory loss that affects cognitive abilities with the aging of individuals, such as language or visual/spatial comprehension. MCI is considered a prodromal phase of more complicated neurodegenerative diseases such as Alzheimer's. Therefore, accurate diagnosis and better understanding of the disease prognosis will facilitate prevention of neurodegeneration. However, the existing diagnostic methods fail to provide precise and well-timed diagnoses, and the pathophysiology of MCI is not fully understood. Alterations of the serum N-glycoproteome expression could represent an essential contributor to the overall pathophysiology of neurodegenerative diseases and be used as a potential marker to assess MCI diagnosis using less invasive procedures. In this approach, we identified N-glycopeptides with different expressions between healthy and MCI patients from serum glycoproteins. Seven of the N-glycopeptides showed outstanding AUC values, among them the antithrombin-III Asn224 + 4-5-0-2 with an AUC value of 1.00 and a p value of 0.0004. According to proteomics and ingenuity pathway analysis (IPA), our data is in line with recent publications, and the glycoproteins carrying the identified N-sites play an important role in neurodegeneration.
Collapse
Affiliation(s)
| | - Atashi Mojgan
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX 79409-1061
| | - Mojibola Fowowe
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX 79409-1061
| | - Sherifdeen Onigbinde
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX 79409-1061
| | - Oluwatosin Daramola
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX 79409-1061
| | - David M. Lubman
- Department of Surgery, The University of Michigan, Ann Arbor, MI 48109
| | - Yehia Mechref
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX 79409-1061
| |
Collapse
|
5
|
Liao J, Chen G, Liu X, Wei ZZ, Yu SP, Chen Q, Ye K. C/EBPβ/AEP signaling couples atherosclerosis to the pathogenesis of Alzheimer's disease. Mol Psychiatry 2022; 27:3034-3046. [PMID: 35422468 PMCID: PMC9912845 DOI: 10.1038/s41380-022-01556-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 03/22/2022] [Accepted: 03/29/2022] [Indexed: 11/08/2022]
Abstract
Atherosclerosis (ATH) and Alzheimer's disease (AD) are both age-dependent inflammatory diseases, associated with infiltrated macrophages and vascular pathology and overlapping molecules. C/EBPβ, an Aβ or inflammatory cytokine-activated transcription factor, and AEP (asparagine endopeptidase) are intimately implicated in both ATH and AD; however, whether C/EBPβ/AEP signaling couples ATH to AD pathogenesis remains incompletely understood. Here we show that C/EBPβ/AEP pathway mediates ATH pathology and couples ATH to AD. Deletion of C/EBPβ or AEP from primary macrophages diminishes cholesterol load, and inactivation of this pathway reduces foam cell formation and lesions in aorta in ApoE-/- mice, fed with HFD (high-fat-diet). Knockout of ApoE from 3xTg AD mouse model augments serum LDL and increases lesion areas in the aorta. Depletion of C/EBPβ or AEP from 3xTg/ApoE-/- mice substantially attenuates these effects and elevates cerebral blood flow and vessel length, improving cognitive functions. Strikingly, knockdown of ApoE from the hippocampus of 3xTg mice decreases the cerebral blood flow and vessel length and aggravates AD pathologies, leading to cognitive deficits. Inactivation of C/EBPβ/AEP pathway alleviates these events and restores cognitive functions. Hence, our findings demonstrate that C/EBPβ/AEP signaling couples ATH to AD via mediating vascular pathology.
Collapse
Affiliation(s)
- Jianming Liao
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Guiqin Chen
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Xia Liu
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Zheng Zachory Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Shan Ping Yu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Qianxue Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| | - Keqiang Ye
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA.
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
6
|
Zhang L, Cao J, Yang H, Pham P, Khan U, Brown B, Wang Y, Zieneldien T, Cao C. Commercial and Instant Coffees Effectively Lower Aβ1-40 and Aβ1-42 in N2a/APPswe Cells. Front Nutr 2022; 9:850523. [PMID: 35369094 PMCID: PMC8965317 DOI: 10.3389/fnut.2022.850523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 02/08/2022] [Indexed: 11/13/2022] Open
Abstract
Background Alzheimer's disease (AD) is a multifactorial neurological disease with neurofibrillary tangles and neuritic plaques as histopathological markers. Due to this, although AD is the leading cause of dementia worldwide, clinical AD dementia cannot be certainly diagnosed until neuropathological post-mortem evaluation. Coffee has been reported to have neurologically protective factors, particularly against AD, but coffee brand and type have not been taken into consideration in previous studies. We examined the discrepancies among popular commercial and instant coffees in limiting the development and progression through Aβ1-40 and Aβ1-42 production, and hypothesized that coffee consumption, regardless of brand or type, is beneficial for stalling the progression and development of Aβ-related AD. Methods Coffee samples from four commercial coffee brands and four instant coffees were purchased or prepared following given instructions and filtered for the study. 5, 2.5, and 1.25% concentrations of each coffee were used to treat N2a/APPswe cell lines. MTT assay was used to assess cell viability for coffee concentrations, as well as pure caffeine concentrations. Sandwich ELISA assay was used to determine Aβ concentration for Aβ1-40 and Aβ1-42 peptides of coffee-treated cells. Results Caffeine concentrations were significantly varied among all coffees (DC vs. MDC, PC, SB, NIN, MIN p < 0.05). There was no correlation between caffeine concentration and cell toxicity among brands and types of coffee, with no toxicity at 0.5 mg/ml caffeine and lower. Most coffees were toxic to N2a/APPswe cells at 5% (p < 0.05), but not at 2.5%. Most coffees at a 2.5% concentration reduced Aβ1-40 and Aβ1-42 production, with comparable results between commercial and instant coffees. Conclusion All coffees tested have beneficial health effects for AD through lowering Aβ1-40 and Aβ1-42 production, with Dunkin' Donuts® medium roast coffee demonstrating the most consistent and optimal cell survival rates and Aβ concentration. On the other hand, Starbucks® coffee exhibited the highest cell toxicity rates among the tested coffees.
Collapse
Affiliation(s)
- Lifang Zhang
- Department of Neurological Rehabilitation, The Affiliated Brain Hospital of Guangzhou Medical University, Guanzhou, China
| | - Jessica Cao
- Department of Kinesiology, Wiess School of Natural Sciences, Rice University, Houston, TX, United States
| | - Haiqiang Yang
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL, United States
| | - Phillip Pham
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL, United States
| | - Umer Khan
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL, United States
| | - Breanna Brown
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL, United States
| | - Yanhong Wang
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL, United States
| | - Tarek Zieneldien
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL, United States
| | - Chuanhai Cao
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL, United States
- Department of Neurology, College of Medicine, University of South Florida, Tampa, FL, United States
| |
Collapse
|
7
|
Domowicz MS, Chan WC, Claudio-Vázquez P, Gonzalez T, Schwartz NB. Brain transcriptome analysis of a CLN2 mouse model as a function of disease progression. J Neuroinflammation 2021; 18:262. [PMID: 34749772 PMCID: PMC8576919 DOI: 10.1186/s12974-021-02302-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 10/20/2021] [Indexed: 12/13/2022] Open
Abstract
Background Neuronal ceroid lipofuscinoses, (NCLs or Batten disease) are a group of inherited, early onset, fatal neurodegenerative diseases associated with mutations in 13 genes. All forms of the disease are characterized by lysosomal accumulation of fluorescent storage material, as well as profound neurodegeneration, but the relationship of the various genes’ function to a single biological process is not obvious. In this study, we used a well-characterized mouse model of classical late infantile NCL (cLINCL) in which the tripeptidyl peptidase 1 (Tpp1) gene is disrupted by gene targeting, resulting in loss of detectable TPP1 activity and leading to progressive neurological phenotypes including ataxia, increased motor deficiency, and early death. Methods In order to identify genes and pathways that may contribute to progression of the neurodegenerative process, we analyzed forebrain/midbrain and cerebellar transcriptional differences at 1, 2, 3 and 4 months of age in control and TPP1-deficient mice by global RNA-sequencing. Results Progressive neurodegenerative inflammatory responses involving microglia, astrocytes and endothelial cells were observed, accompanied by activation of leukocyte extravasation signals and upregulation of nitric oxide production and reactive oxygen species. Several astrocytic (i.e., Gfap, C4b, Osmr, Serpina3n) and microglial (i.e., Ctss, Itgb2, Itgax, Lyz2) genes were identified as strong markers for assessing disease progression as they showed increased levels of expression in vivo over time. Furthermore, transient increased expression of choroid plexus genes was observed at 2 months in the lateral and fourth ventricle, highlighting an early role for the choroid plexus and cerebrospinal fluid in the disease pathology. Based on these gene expression changes, we concluded that neuroinflammation starts, for the most part, after 2 months in the Tpp1−/− brain and that activation of microglia and astrocytes occur more rapidly in cerebellum than in the rest of the brain; confirming increased severity of inflammation in this region. Conclusions These findings have led to a better understanding of cLINCL pathological onset and progression, which may aid in development of future therapeutic treatments for this disease. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02302-z.
Collapse
Affiliation(s)
- Miriam S Domowicz
- Department of Pediatrics, Biological Sciences Division, The University of Chicago Medical Center, 5841 S. Maryland Avenue, MC 5058, Chicago, IL, 60637, USA.
| | - Wen-Ching Chan
- Center for Research Informatics, Biological Sciences Division, The University of Chicago, Chicago, IL, 60637, USA
| | - Patricia Claudio-Vázquez
- Department of Pediatrics, Biological Sciences Division, The University of Chicago Medical Center, 5841 S. Maryland Avenue, MC 5058, Chicago, IL, 60637, USA
| | - Tatiana Gonzalez
- Department of Pediatrics, Biological Sciences Division, The University of Chicago Medical Center, 5841 S. Maryland Avenue, MC 5058, Chicago, IL, 60637, USA
| | - Nancy B Schwartz
- Department of Pediatrics, Biological Sciences Division, The University of Chicago Medical Center, 5841 S. Maryland Avenue, MC 5058, Chicago, IL, 60637, USA.,Department of Biochemistry and Molecular Biology, Biological Sciences Division, The University of Chicago, Chicago, IL, 60637, USA
| |
Collapse
|
8
|
Norton ES, Da Mesquita S, Guerrero-Cazares H. SERPINA3 in glioblastoma and Alzheimer's disease. Aging (Albany NY) 2021; 13:21812-21813. [PMID: 34587119 PMCID: PMC8507276 DOI: 10.18632/aging.203603] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 09/24/2021] [Indexed: 11/25/2022]
Affiliation(s)
- Emily S Norton
- Department of Neurosurgery, Mayo Clinic, Jacksonville, FL 32224, USA.,Neuroscience Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL 32224, USA.,Regenerative Sciences Training Program, Center for Regenerative Medicine, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | - Hugo Guerrero-Cazares
- Department of Neurosurgery, Mayo Clinic, Jacksonville, FL 32224, USA.,Neuroscience Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL 32224, USA.,Regenerative Sciences Training Program, Center for Regenerative Medicine, Mayo Clinic, Jacksonville, FL 32224, USA
| |
Collapse
|
9
|
Iron-responsive-like elements and neurodegenerative ferroptosis. ACTA ACUST UNITED AC 2020; 27:395-413. [PMID: 32817306 PMCID: PMC7433652 DOI: 10.1101/lm.052282.120] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 07/02/2020] [Indexed: 12/26/2022]
Abstract
A set of common-acting iron-responsive 5′untranslated region (5′UTR) motifs can fold into RNA stem loops that appear significant to the biology of cognitive declines of Parkinson's disease dementia (PDD), Lewy body dementia (LDD), and Alzheimer's disease (AD). Neurodegenerative diseases exhibit perturbations of iron homeostasis in defined brain subregions over characteristic time intervals of progression. While misfolding of Aβ from the amyloid-precursor-protein (APP), alpha-synuclein, prion protein (PrP) each cause neuropathic protein inclusions in the brain subregions, iron-responsive-like element (IRE-like) RNA stem–loops reside in their transcripts. APP and αsyn have a role in iron transport while gene duplications elevate the expression of their products to cause rare familial cases of AD and PDD. Of note, IRE-like sequences are responsive to excesses of brain iron in a potential feedback loop to accelerate neuronal ferroptosis and cognitive declines as well as amyloidosis. This pathogenic feedback is consistent with the translational control of the iron storage protein ferritin. We discuss how the IRE-like RNA motifs in the 5′UTRs of APP, alpha-synuclein and PrP mRNAs represent uniquely folded drug targets for therapies to prevent perturbed iron homeostasis that accelerates AD, PD, PD dementia (PDD) and Lewy body dementia, thus preventing cognitive deficits. Inhibition of alpha-synuclein translation is an option to block manganese toxicity associated with early childhood cognitive problems and manganism while Pb toxicity is epigenetically associated with attention deficit and later-stage AD. Pathologies of heavy metal toxicity centered on an embargo of iron export may be treated with activators of APP and ferritin and inhibitors of alpha-synuclein translation.
Collapse
|
10
|
γ-Oryzanol Improves Cognitive Function and Modulates Hippocampal Proteome in Mice. Nutrients 2019; 11:nu11040753. [PMID: 30935111 PMCID: PMC6520752 DOI: 10.3390/nu11040753] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 03/27/2019] [Accepted: 03/28/2019] [Indexed: 01/18/2023] Open
Abstract
Rice (Oryza sativa L.) is the richest source of γ-oryzanol, a compound endowed with antioxidant and anti-inflammatory properties. γ-Oryzanol has been demonstrated to cross the blood-brain barrier in intact form and exert beneficial effects on brain function. This study aimed to clarify the effects of γ-oryzanol in the hippocampus in terms of cognitive function and protein expression. Adult mice were administered with γ-oryzanol 100 mg/kg or vehicle (control) once a day for 21 consecutive days following which cognitive behavior and hippocampal proteome were investigated. Cognitive tests using novel object recognition and Y-maze showed that long-term consumption of γ-oryzanol improves cognitive function in mice. To investigate the hippocampal proteome modulated by γ-oryzanol, 2D-difference gel electrophoresis (2D-DIGE) was performed. Interestingly, we found that γ-oryzanol modulates quantitative changes of proteins involved in synaptic plasticity and neuronal trafficking, neuroprotection and antioxidant activity, and mitochondria and energy metabolism. These findings suggested γ-oryzanol as a natural compound able to maintain and reinforce brain function. Although more intensive studies are needed, we propose γ-oryzanol as a putative dietary phytochemical for preserving brain reserve, the ability to tolerate age-related changes, thereby preventing clinical symptoms or signs of neurodegenerative diseases.
Collapse
|
11
|
Karthick C, Nithiyanandan S, Essa MM, Guillemin GJ, Jayachandran SK, Anusuyadevi M. Time-dependent effect of oligomeric amyloid-β (1-42)-induced hippocampal neurodegeneration in rat model of Alzheimer's disease. Neurol Res 2018; 41:139-150. [PMID: 30453864 DOI: 10.1080/01616412.2018.1544745] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Alzheimer's disease (AD) is characterized with an abnormal deposition of insoluble amyloid-beta (Aβ) peptide plaques, tangles formation and synaptic dysfunction. These result in impaired functioning of neuronal circuits and alter the behavioral response owing to activation of neurotransmitter receptors. Recently, it has been implicated that Aβ influences N-methyl d-aspartate (NMDA) receptor activation in AD; however, the molecular mechanism underlying remains unclear. Thus, emerged specific aim to study the time-course effect of oligomeric Aβ(1-42) (oAβ1-42) on the mRNA expression of genes encoding NMDA and acetylcholine receptors in the rat model of AD. METHODS Aggregated forms of synthetic Aβ peptides were injected bilaterally into the intrahippocampal region of rat brain using stereotaxic surgery. Behavioral analysis was performed using eight-arm Radial Arm Maze task at the end of experimental period. Euthanized rat brain hippocampal tissue was used to study the mRNA expression of glutamatergic and cholinergic receptor using semiquantitative reverse transcription-polymerase chain reaction. RESULTS oAβ1-42 decreased the gene expression level of α7-nicotinic acetylcholine receptor and increased the mRNA expression of NMDA receptor 2A, and -2B subunits. In particular, oAβ1-42 aggregates increased the retention time and altered the behavioral response in rats after 15 days of injection. Further, amyloid-β1-42 are highly expressed in 15 days after postinjection in hippocampus of adult rats. CONCLUSION Acute exposure of oAβ1-42 modulated differential gene expression of glutamatergic and cholinergic receptors in hippocampus of adult rats and is duration dependent reflecting changes in hippocampal circuitry system underlying learning and memory impairments. ABBREVIATIONS AD: Alzheimer's disease, Aβ: amyloid-β; oAβ1-42: oligomeric amyloid-β 1-42 full length peptide; CAM: calmodulin; CNS: central nervous system; CR: Congo red; DG: dentate gyrus; EC: entorhinal cortex; HFIP: 1,1,1,3,3,3-hexafluoro-2-propanol; IBO: ibotenic acid; NMDA: N-methyl d-aspartate; NMDAR: N-methyl d-aspartate receptor; NR2A: N-methyl d-aspartate receptor 2A; NR2B: N-methyl d-aspartate receptor 2B; ACh: acetylcholine; α7-nAChR: α7-nicotinic acetylcholine receptor; PBS: phosphate buffered saline; RAM: Radial Arm Maze; ThT: thioflavin T.
Collapse
Affiliation(s)
- Chennakesavan Karthick
- a Molecular Gerontology Laboratory, Department of Biochemistry , School of Life Sciences, Bharathidasan University , Tiruchirappalli , India
| | - Saravanan Nithiyanandan
- a Molecular Gerontology Laboratory, Department of Biochemistry , School of Life Sciences, Bharathidasan University , Tiruchirappalli , India
| | - Musthafa Mohamed Essa
- b Department of Food Science and Nutrition , College of Agriculture and Marine Sciences, Sultan Qaboos University , Muscat , Oman
| | - Gilles J Guillemin
- c Neuroinflammation group, Faculty of Medicine and Health Sciences, Deb Bailey MND Research Laboratory , Macquaire University , Sydney , Australia
| | - Swaminathan K Jayachandran
- d Molecular Cardiology and Drug Discovery Laboratory, Department of Bioinformatics , School of Life Sciences, Bharathidasan University , Tiruchirappalli , India
| | - Muthuswamy Anusuyadevi
- a Molecular Gerontology Laboratory, Department of Biochemistry , School of Life Sciences, Bharathidasan University , Tiruchirappalli , India
| |
Collapse
|
12
|
Yang J, Zhang R, Shi C, Mao C, Yang Z, Suo Z, Torp R, Xu Y. AQP4 Association with Amyloid Deposition and Astrocyte Pathology in the Tg-ArcSwe Mouse Model of Alzheimer's Disease. J Alzheimers Dis 2018; 57:157-169. [PMID: 28222512 DOI: 10.3233/jad-160957] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Amyloid-β deposition in senile plaques is one of the main pathological changes in Alzheimer's disease (AD). We previously reported that aquaporin-4 (AQP4) is redistributed within the astrocytes in cerebral amyloid angiopathy in the tg-ArcSwe mouse model of AD, suggesting that AQP4 may participate in amyloid-β deposition. However, the role of AQP4 in plaque formation is not currently clear. The objective of the current study was to explore the AQP4 distribution within plaques in the tg-ArcSwe mice in more depth by the combined application of immunofluorescence cytochemistry and immunogold electron microscopy. In addition, the astrocyte marker, glial fibrillary acidic protein (GFAP), was studied in association with AQP4. We demonstrated a robust upregulation of AQP4 expression in areas of plaques. Compared to GFAP, AQP4 appeared predominantly at later stages of plaque formation, in older mice, and within the processes of astrocytes. In combination with GFAP, AQP4 differentiated plaques into three progression stages under light microscopy. This suggests that AQP4 expression was associated with amyloid deposition and astrocyte pathology in the Tg-ArcSwe mouse model of AD. This provides novel proof for the involvement of AQP4 in the process of amyloid deposition in AD.
Collapse
Affiliation(s)
- Jing Yang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Henan, China
| | - Rui Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Henan, China
| | - Changhe Shi
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Henan, China
| | - Chengyuan Mao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Henan, China
| | - Zhihua Yang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Henan, China
| | - Zhenhe Suo
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway.,Department of Pathology, The Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Reidun Torp
- Department of Anatomy, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Yuming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Henan, China
| |
Collapse
|
13
|
Sukoff Rizzo SJ, Crawley JN. Behavioral Phenotyping Assays for Genetic Mouse Models of Neurodevelopmental, Neurodegenerative, and Psychiatric Disorders. Annu Rev Anim Biosci 2017; 5:371-389. [PMID: 28199172 DOI: 10.1146/annurev-animal-022516-022754] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Animal models offer heuristic research tools to understand the causes of human diseases and to identify potential treatments. With rapidly evolving genetic engineering technologies, mutations identified in a human disorder can be generated in the mouse genome. Phenotypic outcomes of the mutation are then explicated to confirm hypotheses about causes and to discover effective therapeutics. Most neurodevelopmental, neurodegenerative, and psychiatric disorders are diagnosed primarily by their prominent behavioral symptoms. Mouse behavioral assays analogous to the human symptoms have been developed to analyze the consequences of mutations and to evaluate proposed therapeutics preclinically. Here we describe the range of mouse behavioral tests available in the established behavioral neuroscience literature, along with examples of their translational applications. Concepts presented have been successfully used in other species, including flies, worms, fish, rats, pigs, and nonhuman primates. Identical strategies can be employed to test hypotheses about environmental causes and gene × environment interactions.
Collapse
Affiliation(s)
| | - Jacqueline N Crawley
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California, Davis School of Medicine, Sacramento, California 95817;
| |
Collapse
|
14
|
Tai LM, Balu D, Avila-Munoz E, Abdullah L, Thomas R, Collins N, Valencia-Olvera AC, LaDu MJ. EFAD transgenic mice as a human APOE relevant preclinical model of Alzheimer's disease. J Lipid Res 2017; 58:1733-1755. [PMID: 28389477 PMCID: PMC5580905 DOI: 10.1194/jlr.r076315] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 04/06/2017] [Indexed: 01/12/2023] Open
Abstract
Identified in 1993, APOE4 is the greatest genetic risk factor for sporadic Alzheimer's disease (AD), increasing risk up to 15-fold compared with APOE3, with APOE2 decreasing AD risk. However, the functional effects of APOE4 on AD pathology remain unclear and, in some cases, controversial. In vivo progress to understand how the human (h)-APOE genotypes affect AD pathology has been limited by the lack of a tractable familial AD-transgenic (FAD-Tg) mouse model expressing h-APOE rather than mouse (m)-APOE. The disparity between m- and h-apoE is relevant for virtually every AD-relevant pathway, including amyloid-β (Aβ) deposition and clearance, neuroinflammation, tau pathology, neural plasticity and cerebrovascular deficits. EFAD mice were designed as a temporally useful preclinical FAD-Tg-mouse model expressing the h-APOE genotypes for identifying mechanisms underlying APOE-modulated symptoms of AD pathology. From their first description in 2012, EFAD mice have enabled critical basic and therapeutic research. Here we review insights gleaned from the EFAD mice and summarize future directions.
Collapse
Affiliation(s)
- Leon M Tai
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612
| | - Deebika Balu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612
| | - Evangelina Avila-Munoz
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612
| | | | - Riya Thomas
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612
| | - Nicole Collins
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612
| | | | - Mary Jo LaDu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612.
| |
Collapse
|
15
|
Andreasson KI, Bachstetter AD, Colonna M, Ginhoux F, Holmes C, Lamb B, Landreth G, Lee DC, Low D, Lynch MA, Monsonego A, O’Banion MK, Pekny M, Puschmann T, Russek-Blum N, Sandusky LA, Selenica MLB, Takata K, Teeling J, Town T, Van Eldik LJ, Russek-Blum N, Monsonego A, Low D, Takata K, Ginhoux F, Town T, O’Banion MK, Lamb B, Colonna M, Landreth G, Andreasson KI, Sandusky LA, Selenica MLB, Lee DC, Holmes C, Teeling J, Lynch MA, Van Eldik LJ, Bachstetter AD, Pekny M, Puschmann T. Targeting innate immunity for neurodegenerative disorders of the central nervous system. J Neurochem 2016; 138:653-93. [PMID: 27248001 PMCID: PMC5433264 DOI: 10.1111/jnc.13667] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 04/01/2016] [Accepted: 04/30/2016] [Indexed: 12/21/2022]
Abstract
Neuroinflammation is critically involved in numerous neurodegenerative diseases, and key signaling steps of innate immune activation hence represent promising therapeutic targets. This mini review series originated from the 4th Venusberg Meeting on Neuroinflammation held in Bonn, Germany, 7-9th May 2015, presenting updates on innate immunity in acute brain injury and chronic neurodegenerative disorders, such as traumatic brain injury and Alzheimer disease, on the role of astrocytes and microglia, as well as technical developments that may help elucidate neuroinflammatory mechanisms and establish clinical relevance. In this meeting report, a brief overview of physiological and pathological microglia morphology is followed by a synopsis on PGE2 receptors, insights into the role of arginine metabolism and further relevant aspects of neuroinflammation in various clinical settings, and concluded by a presentation of technical challenges and solutions when working with microglia and astrocyte cultures. Microglial ontogeny and induced pluripotent stem cell-derived microglia, advances of TREM2 signaling, and the cytokine paradox in Alzheimer's disease are further contributions to this article. Neuroinflammation is critically involved in numerous neurodegenerative diseases, and key signaling steps of innate immune activation hence represent promising therapeutic targets. This mini review series originated from the 4th Venusberg Meeting on Neuroinflammation held in Bonn, Germany, 7-9th May 2015, presenting updates on innate immunity in acute brain injury and chronic neurodegenerative disorders, such as traumatic brain injury and Alzheimer's disease, on the role of astrocytes and microglia, as well as technical developments that may help elucidate neuroinflammatory mechanisms and establish clinical relevance. In this meeting report, a brief overview on physiological and pathological microglia morphology is followed by a synopsis on PGE2 receptors, insights into the role of arginine metabolism and further relevant aspects of neuroinflammation in various clinical settings, and concluded by a presentation of technical challenges and solutions when working with microglia cultures. Microglial ontogeny and induced pluripotent stem cell-derived microglia, advances of TREM2 signaling, and the cytokine paradox in Alzheimer's disease are further contributions to this article.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Niva Russek-Blum
- The Dead Sea and Arava Science Center, Central Arava Branch, Yair Station, Hazeva, Israel
| | - Alon Monsonego
- The Shraga Segal Dept. of Microbiology, Immunology and Genetics, The Faculty of Health Sciences: The National Institute of Biotechnology in the Negev, and Zlotowski Center for Neuroscience, Ben-Gurion University, Beer-Sheva 84105, Israel
| | - Donovan Low
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Kazuyuki Takata
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
- Department of Clinical and Translational Physiology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Terrence Town
- Departments of Physiology and Biophysics, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089,
| | - M. Kerry O’Banion
- Departments of Neuroscience and Neurology, Del Monte Neuromedicine Institute, University of Rochester School of Medicine & Dentistry, Rochester, NY 14642,
| | - Bruce Lamb
- Department of Neurosciences, Cleveland Clinic, Cleveland, OH 44106
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Gary Landreth
- Department of Neurosciences, Case Western Reserve University 44106
| | - Katrin I. Andreasson
- Department of Neurology and Neurological Sciences, Stanford Neuroscience Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Leslie A. Sandusky
- USF Health Byrd Alzheimer’s Institute, Tampa, FL 33613
- College of Pharmacy & Pharmaceutical Sciences, Tampa, FL 33613
| | - Maj-Linda B. Selenica
- USF Health Byrd Alzheimer’s Institute, Tampa, FL 33613
- College of Pharmacy & Pharmaceutical Sciences, Tampa, FL 33613
| | - Daniel C. Lee
- USF Health Byrd Alzheimer’s Institute, Tampa, FL 33613
- College of Pharmacy & Pharmaceutical Sciences, Tampa, FL 33613
| | - Clive Holmes
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Tremona Road, Southampton, SO16 7YD, United Kingdom
| | - Jessica Teeling
- Centre for Biological Sciences, University of Southampton, Southampton General Hospital, Tremona Road, Southampton, SO16 7YD, United Kingdom
| | | | | | | | - Milos Pekny
- Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, SE-405 30 Gothenburg, Sweden
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
- Hunter Medical Research Institute, University of Newcastle, New South Wales, Australia
| | - Till Puschmann
- Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, SE-405 30 Gothenburg, Sweden
| |
Collapse
|
16
|
Faucher P, Mons N, Micheau J, Louis C, Beracochea DJ. Hippocampal Injections of Oligomeric Amyloid β-peptide (1-42) Induce Selective Working Memory Deficits and Long-lasting Alterations of ERK Signaling Pathway. Front Aging Neurosci 2016; 7:245. [PMID: 26793098 PMCID: PMC4707555 DOI: 10.3389/fnagi.2015.00245] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 12/14/2015] [Indexed: 12/24/2022] Open
Abstract
Increasing evidence suggests that abnormal brain accumulation of soluble rather than aggregated amyloid-β1-42 oligomers (Aβo(1-42)) plays a causal role in Alzheimer's disease (AD). However, as yet, animal's models of AD based on oligomeric amyloid-β1-42 injections in the brain have not investigated their long-lasting impacts on molecular and cognitive functions. In addition, the injections have been most often performed in ventricles, but not in the hippocampus, in spite of the fact that the hippocampus is importantly involved in memory processes and is strongly and precociously affected during the early stages of AD. Thus, in the present study, we investigated the long-lasting impacts of intra-hippocampal injections of oligomeric forms of Aβo(1-42) on working and spatial memory and on the related activation of ERK1/2. Indeed, the extracellular signal-regulated kinase (ERK) which is involved in memory function had been found to be activated by amyloid peptides. We found that repeated bilateral injections (1injection/day over 4 successive days) of oligomeric forms of Aβo(1-42) into the dorsal hippocampus lead to long-lasting impairments in two working memory tasks, these deficits being observed 7 days after the last injection, while spatial memory remained unaffected. Moreover, the working memory deficits were correlated with sustained impairments of ERK1/2 activation in the medial prefrontal cortex (mPFC) and the septum, two brain areas tightly connected with the hippocampus and involved in working memory. Thus, our study is first to evidence that sub-chronic injections of oligomeric forms of Aβo(1-42) into the dorsal hippocampus produces the main sign of cognitive impairments corresponding to the early stages of AD, via long-lasting alterations of an ERK/MAPK pathway in an interconnected brain networks.
Collapse
Affiliation(s)
- Pierre Faucher
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, Université de Bordeaux, CNRS, UMR 5287 Pessac, France
| | - Nicole Mons
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, Université de Bordeaux, CNRS, UMR 5287 Pessac, France
| | - Jacques Micheau
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, Université de Bordeaux, CNRS, UMR 5287 Pessac, France
| | - Caroline Louis
- Institut de Recherches Servier Croissy sur Seine, France
| | - Daniel J Beracochea
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, Université de Bordeaux, CNRS, UMR 5287 Pessac, France
| |
Collapse
|
17
|
Chakrabarty P, Li A, Ceballos-Diaz C, Eddy JA, Funk CC, Moore B, DiNunno N, Rosario AM, Cruz PE, Verbeeck C, Sacino A, Nix S, Janus C, Price ND, Das P, Golde TE. IL-10 alters immunoproteostasis in APP mice, increasing plaque burden and worsening cognitive behavior. Neuron 2015; 85:519-33. [PMID: 25619653 DOI: 10.1016/j.neuron.2014.11.020] [Citation(s) in RCA: 277] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2013] [Revised: 10/24/2014] [Accepted: 11/20/2014] [Indexed: 01/27/2023]
Abstract
Anti-inflammatory strategies are proposed to have beneficial effects in Alzheimer's disease. To explore how anti-inflammatory cytokine signaling affects Aβ pathology, we investigated the effects of adeno-associated virus (AAV2/1)-mediated expression of Interleukin (IL)-10 in the brains of APP transgenic mouse models. IL-10 expression resulted in increased Aβ accumulation and impaired memory in APP mice. A focused transcriptome analysis revealed changes consistent with enhanced IL-10 signaling and increased ApoE expression in IL-10-expressing APP mice. ApoE protein was selectively increased in the plaque-associated insoluble cellular fraction, likely because of direct interaction with aggregated Aβ in the IL-10-expressing APP mice. Ex vivo studies also show that IL-10 and ApoE can individually impair glial Aβ phagocytosis. Our observations that IL-10 has an unexpected negative effect on Aβ proteostasis and cognition in APP mouse models demonstrate the complex interplay between innate immunity and proteostasis in neurodegenerative diseases, an interaction we call immunoproteostasis.
Collapse
Affiliation(s)
- Paramita Chakrabarty
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA.
| | - Andrew Li
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Carolina Ceballos-Diaz
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - James A Eddy
- Institute for Systems Biology, 401 Terry Avenue N, Seattle, WA 98109, USA
| | - Cory C Funk
- Institute for Systems Biology, 401 Terry Avenue N, Seattle, WA 98109, USA
| | - Brenda Moore
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Nadia DiNunno
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Awilda M Rosario
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Pedro E Cruz
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Christophe Verbeeck
- Department of Neuroscience, Mayo Clinic College of Medicine, Jacksonville, FL 32224, USA
| | - Amanda Sacino
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Sarah Nix
- Department of Neuroscience, Mayo Clinic College of Medicine, Jacksonville, FL 32224, USA
| | - Christopher Janus
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Nathan D Price
- Institute for Systems Biology, 401 Terry Avenue N, Seattle, WA 98109, USA
| | - Pritam Das
- Department of Neuroscience, Mayo Clinic College of Medicine, Jacksonville, FL 32224, USA
| | - Todd E Golde
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
18
|
Huang Y, Mahley RW. Apolipoprotein E: structure and function in lipid metabolism, neurobiology, and Alzheimer's diseases. Neurobiol Dis 2014; 72 Pt A:3-12. [PMID: 25173806 PMCID: PMC4253862 DOI: 10.1016/j.nbd.2014.08.025] [Citation(s) in RCA: 505] [Impact Index Per Article: 45.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 08/06/2014] [Accepted: 08/20/2014] [Indexed: 12/01/2022] Open
Abstract
Apolipoprotein (apo) E is a multifunctional protein with central roles in lipid metabolism, neurobiology, and neurodegenerative diseases. It has three major isoforms (apoE2, apoE3, and apoE4) with different effects on lipid and neuronal homeostasis. A major function of apoE is to mediate the binding of lipoproteins or lipid complexes in the plasma or interstitial fluids to specific cell-surface receptors. These receptors internalize apoE-containing lipoprotein particles; thus, apoE participates in the distribution/redistribution of lipids among various tissues and cells of the body. In addition, intracellular apoE may modulate various cellular processes physiologically or pathophysiologically, including cytoskeletal assembly and stability, mitochondrial integrity and function, and dendritic morphology and function. Elucidation of the functional domains within this protein and of the three-dimensional structure of the major isoforms of apoE has contributed significantly to our understanding of its physiological and pathophysiological roles at a molecular level. It is likely that apoE, with its multiple cellular origins and multiple structural and biophysical properties, is involved widely in processes of lipid metabolism and neurobiology, possibly encompassing a variety of disorders of neuronal repair, remodeling, and degeneration by interacting with different factors through various pathways.
Collapse
Affiliation(s)
- Yadong Huang
- Gladstone Institute of Neurological Disease, University of California, San Francisco 94158, USA; Gladstone Institute of Cardiovascular Disease, University of California, San Francisco 94158, USA; Department of Neurology, University of California, San Francisco 94158, USA; Department of Pathology, University of California, San Francisco 94158, USA.
| | - Robert W Mahley
- Gladstone Institute of Neurological Disease, University of California, San Francisco 94158, USA; Gladstone Institute of Cardiovascular Disease, University of California, San Francisco 94158, USA; Department of Pathology, University of California, San Francisco 94158, USA; Department of Medicine, University of California, San Francisco 94158, USA
| |
Collapse
|
19
|
Webster SJ, Bachstetter AD, Nelson PT, Schmitt FA, Van Eldik LJ. Using mice to model Alzheimer's dementia: an overview of the clinical disease and the preclinical behavioral changes in 10 mouse models. Front Genet 2014; 5:88. [PMID: 24795750 PMCID: PMC4005958 DOI: 10.3389/fgene.2014.00088] [Citation(s) in RCA: 513] [Impact Index Per Article: 46.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 04/01/2014] [Indexed: 01/17/2023] Open
Abstract
The goal of this review is to discuss how behavioral tests in mice relate to the pathological and neuropsychological features seen in human Alzheimer's disease (AD), and present a comprehensive analysis of the temporal progression of behavioral impairments in commonly used AD mouse models that contain mutations in amyloid precursor protein (APP). We begin with a brief overview of the neuropathological changes seen in the AD brain and an outline of some of the clinical neuropsychological assessments used to measure cognitive deficits associated with the disease. This is followed by a critical assessment of behavioral tasks that are used in AD mice to model the cognitive changes seen in the human disease. Behavioral tests discussed include spatial memory tests [Morris water maze (MWM), radial arm water maze (RAWM), Barnes maze], associative learning tasks (passive avoidance, fear conditioning), alternation tasks (Y-Maze/T-Maze), recognition memory tasks (Novel Object Recognition), attentional tasks (3 and 5 choice serial reaction time), set-shifting tasks, and reversal learning tasks. We discuss the strengths and weaknesses of each of these behavioral tasks, and how they may correlate with clinical assessments in humans. Finally, the temporal progression of both cognitive and non-cognitive deficits in 10 AD mouse models (PDAPP, TG2576, APP23, TgCRND8, J20, APP/PS1, TG2576 + PS1 (M146L), APP/PS1 KI, 5×FAD, and 3×Tg-AD) are discussed in detail. Mouse models of AD and the behavioral tasks used in conjunction with those models are immensely important in contributing to our knowledge of disease progression and are a useful tool to study AD pathophysiology and the resulting cognitive deficits. However, investigators need to be aware of the potential weaknesses of the available preclinical models in terms of their ability to model cognitive changes observed in human AD. It is our hope that this review will assist investigators in selecting an appropriate mouse model, and accompanying behavioral paradigms to investigate different aspects of AD pathology and disease progression.
Collapse
Affiliation(s)
- Scott J Webster
- Sanders-Brown Center on Aging, University of Kentucky Lexington, KY, USA
| | - Adam D Bachstetter
- Sanders-Brown Center on Aging, University of Kentucky Lexington, KY, USA
| | - Peter T Nelson
- Sanders-Brown Center on Aging, University of Kentucky Lexington, KY, USA ; Division of Neuropathology, Department of Pathology and Laboratory Medicine, University of Kentucky Lexington, KY, USA
| | - Frederick A Schmitt
- Sanders-Brown Center on Aging, University of Kentucky Lexington, KY, USA ; Department of Neurology, University of Kentucky Lexington, KY, USA
| | - Linda J Van Eldik
- Sanders-Brown Center on Aging, University of Kentucky Lexington, KY, USA ; Department of Anatomy and Neurobiology, University of Kentucky Lexington, KY, USA
| |
Collapse
|
20
|
Lathe R, Sapronova A, Kotelevtsev Y. Atherosclerosis and Alzheimer--diseases with a common cause? Inflammation, oxysterols, vasculature. BMC Geriatr 2014; 14:36. [PMID: 24656052 PMCID: PMC3994432 DOI: 10.1186/1471-2318-14-36] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 02/26/2014] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Aging is accompanied by increasing vulnerability to pathologies such as atherosclerosis (ATH) and Alzheimer disease (AD). Are these different pathologies, or different presentations with a similar underlying pathoetiology? DISCUSSION Both ATH and AD involve inflammation, macrophage infiltration, and occlusion of the vasculature. Allelic variants in common genes including APOE predispose to both diseases. In both there is strong evidence of disease association with viral and bacterial pathogens including herpes simplex and Chlamydophila. Furthermore, ablation of components of the immune system (or of bone marrow-derived macrophages alone) in animal models restricts disease development in both cases, arguing that both are accentuated by inflammatory/immune pathways. We discuss that amyloid β, a distinguishing feature of AD, also plays a key role in ATH. Several drugs, at least in mouse models, are effective in preventing the development of both ATH and AD. Given similar age-dependence, genetic underpinnings, involvement of the vasculature, association with infection, Aβ involvement, the central role of macrophages, and drug overlap, we conclude that the two conditions reflect different manifestations of a common pathoetiology. MECHANISM Infection and inflammation selectively induce the expression of cholesterol 25-hydroxylase (CH25H). Acutely, the production of 'immunosterol' 25-hydroxycholesterol (25OHC) defends against enveloped viruses. We present evidence that chronic macrophage CH25H upregulation leads to catalyzed esterification of sterols via 25OHC-driven allosteric activation of ACAT (acyl-CoA cholesterol acyltransferase/SOAT), intracellular accumulation of cholesteryl esters and lipid droplets, vascular occlusion, and overt disease. SUMMARY We postulate that AD and ATH are both caused by chronic immunologic challenge that induces CH25H expression and protection against particular infectious agents, but at the expense of longer-term pathology.
Collapse
Affiliation(s)
- Richard Lathe
- State University of Pushchino, Prospekt Nauki, Pushchino 142290, Moscow Region, Russia
- Pushchino Branch of the Institute of Bioorganic Chemistry, Russian Academy of Sciences, Pushchino 142290 Moscow Region, Russia
- Pieta Research, PO Box 27069, Edinburgh EH10 5YW, UK
| | - Alexandra Sapronova
- State University of Pushchino, Prospekt Nauki, Pushchino 142290, Moscow Region, Russia
- Pushchino Branch of the Institute of Bioorganic Chemistry, Russian Academy of Sciences, Pushchino 142290 Moscow Region, Russia
- Optical Research Group, Laboratory of Evolutionary Biophysics of Development, Institute of Developmental Biology of the Russian Academy of Sciences, Moscow, Russia
| | - Yuri Kotelevtsev
- State University of Pushchino, Prospekt Nauki, Pushchino 142290, Moscow Region, Russia
- Pushchino Branch of the Institute of Bioorganic Chemistry, Russian Academy of Sciences, Pushchino 142290 Moscow Region, Russia
- Biomedical Centre for Research Education and Innovation (CREI), Skolkovo Institute of Science and Technology, Skolkovo 143025, Russia
- Centre for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Little France, Edinburgh EH16 4TJ, UK
| |
Collapse
|
21
|
Klohs J, Rudin M, Shimshek DR, Beckmann N. Imaging of cerebrovascular pathology in animal models of Alzheimer's disease. Front Aging Neurosci 2014; 6:32. [PMID: 24659966 PMCID: PMC3952109 DOI: 10.3389/fnagi.2014.00032] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 02/19/2014] [Indexed: 01/04/2023] Open
Abstract
In Alzheimer's disease (AD), vascular pathology may interact with neurodegeneration and thus aggravate cognitive decline. As the relationship between these two processes is poorly understood, research has been increasingly focused on understanding the link between cerebrovascular alterations and AD. This has at last been spurred by the engineering of transgenic animals, which display pathological features of AD and develop cerebral amyloid angiopathy to various degrees. Transgenic models are versatile for investigating the role of amyloid deposition and vascular dysfunction, and for evaluating novel therapeutic concepts. In addition, research has benefited from the development of novel imaging techniques, which are capable of characterizing vascular pathology in vivo. They provide vascular structural read-outs and have the ability to assess the functional consequences of vascular dysfunction as well as to visualize and monitor the molecular processes underlying these pathological alterations. This article focusses on recent in vivo small animal imaging studies addressing vascular aspects related to AD. With the technical advances of imaging modalities such as magnetic resonance, nuclear and microscopic imaging, molecular, functional and structural information related to vascular pathology can now be visualized in vivo in small rodents. Imaging vascular and parenchymal amyloid-β (Aβ) deposition as well as Aβ transport pathways have been shown to be useful to characterize their dynamics and to elucidate their role in the development of cerebral amyloid angiopathy and AD. Structural and functional imaging read-outs have been employed to describe the deleterious affects of Aβ on vessel morphology, hemodynamics and vascular integrity. More recent imaging studies have also addressed how inflammatory processes partake in the pathogenesis of the disease. Moreover, imaging can be pivotal in the search for novel therapies targeting the vasculature.
Collapse
Affiliation(s)
- Jan Klohs
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich Zurich, Switzerland ; Neuroscience Center Zurich, University of Zurich and ETH Zurich Zurich, Switzerland
| | - Markus Rudin
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich Zurich, Switzerland ; Neuroscience Center Zurich, University of Zurich and ETH Zurich Zurich, Switzerland ; Institute of Pharmacology and Toxicology, University of Zurich Zurich, Switzerland
| | - Derya R Shimshek
- Autoimmunity, Transplantation and Inflammation/Neuroinflammation Department, Novartis Institutes for BioMedical Research Basel, Switzerland
| | - Nicolau Beckmann
- Analytical Sciences and Imaging, Novartis Institutes for BioMedical Research Basel, Switzerland
| |
Collapse
|
22
|
Sasaoka N, Sakamoto M, Kanemori S, Kan M, Tsukano C, Takemoto Y, Kakizuka A. Long-term oral administration of hop flower extracts mitigates Alzheimer phenotypes in mice. PLoS One 2014; 9:e87185. [PMID: 24489866 PMCID: PMC3906130 DOI: 10.1371/journal.pone.0087185] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Accepted: 12/23/2013] [Indexed: 01/08/2023] Open
Abstract
Coincident with the expanding population of aged people, the incidence of Alzheimer disease (AD) is rapidly increasing in most advanced countries. At present, no effective prophylactics are available. Among several pathological mechanisms proposed for AD, the “amyloid hypothesis” has been most widely accepted, in which accumulation or deposition of Aβ is considered to be the initial event. Thus, prevention of Aβ production would be an ideal strategy for the treatment or prevention of AD. Aβ is produced via the proteolytic cleavage of its precursor protein, APP (amyloid precursor protein), by two different enzymes, β and γ-secretases. Indeed, inhibitors against either or both enzymes have been developed and tested for clinical efficacy. Based on the “amyloid hypothesis”, we developed a luciferase-based screening method to monitor γ-secretase activity, screened more than 1,600 plant extracts, most of which have long been used in Chinese medicine, and observed that Hop extracts significantly inhibit Aβ production in cultured cells. A major component of the inhibitory activity was purified, and its chemical identity was determined by NMR to be Garcinielliptone HC. In vivo, oral administration of Hop extracts to AD model mice decreased Aβ depositions in the cerebral cortex of the parietal lobe, hippocampus, and artery walls (amyloid angiopathy) in the brains. In a Morris water maze test, AD model mice that had daily consumed Hop extracts in their drinking water showed significant mitigation of memory impairment at ages of 9 and 12 months. Moreover, in the open field test oral administration of Hop extracts also prevented an emotional disturbance that appeared in the AD mice at 18 months. Despite lifelong consumption of Hop extracts, no deleterious side effects were observed at any age. These results support the “amyloid hypothesis”, and indicate that Hop extract is a promising candidate for an effective prophylactic for AD.
Collapse
Affiliation(s)
- Norio Sasaoka
- Laboratory of Functional Biology, Kyoto University Graduate School of Biostudies, Sakyo-ku, Kyoto, Japan
| | - Megumi Sakamoto
- Laboratory of Functional Biology, Kyoto University Graduate School of Biostudies, Sakyo-ku, Kyoto, Japan
| | - Shoko Kanemori
- Laboratory of Functional Biology, Kyoto University Graduate School of Biostudies, Sakyo-ku, Kyoto, Japan
| | - Michiru Kan
- Laboratory of Functional Biology, Kyoto University Graduate School of Biostudies, Sakyo-ku, Kyoto, Japan
| | - Chihiro Tsukano
- Department of Organic Chemistry, Kyoto University Graduate School of Pharmaceutical Sciences, Sakyo-ku, Kyoto, Japan
| | - Yoshiji Takemoto
- Department of Organic Chemistry, Kyoto University Graduate School of Pharmaceutical Sciences, Sakyo-ku, Kyoto, Japan
| | - Akira Kakizuka
- Laboratory of Functional Biology, Kyoto University Graduate School of Biostudies, Sakyo-ku, Kyoto, Japan
- * E-mail:
| |
Collapse
|
23
|
Alpha 1-Antichymotrypsin, an Inflammatory Protein Overexpressed in the Brains of Patients with Alzheimer's Disease, Induces Tau Hyperphosphorylation through c-Jun N-Terminal Kinase Activation. Int J Alzheimers Dis 2013; 2013:606083. [PMID: 24175110 PMCID: PMC3794555 DOI: 10.1155/2013/606083] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Revised: 08/08/2013] [Accepted: 08/13/2013] [Indexed: 01/09/2023] Open
Abstract
The association of inflammatory proteins with neuritic plaques in the brains of Alzheimer's disease (AD) patients has led to the hypothesis that inflammation plays a pivotal role in the development of pathology in AD. Earlier studies have shown that alpha 1-antichymotrypsin (ACT) enhances amyloid beta fibrillization and accelerated plaque formation in APP transgenic mice. Later studies from our laboratory have shown that purified ACT induces tau hyperphosphorylation and degeneration in neurons. In order to understand the mechanisms by which inflammatory proteins enhance tau hyperphosphorylation, we injected interleukin-1β (IL-1β) intracerebroventricularly into mice expressing human ACT, human tau, or both transgenes. It was found that the hyperphosphorylation of tau in ACT and ACT/htau mice after IL-1β injection correlated with increased phosphorylation of c-Jun N-terminal kinase (JNK). We verified the involvement of JNK in ACT-induced tau phosphorylation by utilizing JNK inhibitors in cultured primary neurons treated with ACT, and we found that the inhibitor showed complete prevention of ACT-induced tau phosphorylation. These results indicate that JNK is one of the major kinases involved in the ACT-mediated tau hyperphosphorylation and suggest that inhibitors of this kinase may protect against inflammation-induced tau hyperphosphorylation and neurodegeneration associated with AD.
Collapse
|
24
|
Bandyopadhyay S, Cahill C, Balleidier A, Huang C, Lahiri DK, Huang X, Rogers JT. Novel 5' untranslated region directed blockers of iron-regulatory protein-1 dependent amyloid precursor protein translation: implications for down syndrome and Alzheimer's disease. PLoS One 2013; 8:e65978. [PMID: 23935819 PMCID: PMC3729844 DOI: 10.1371/journal.pone.0065978] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2013] [Accepted: 04/30/2013] [Indexed: 11/19/2022] Open
Abstract
We reported that iron influx drives the translational expression of the neuronal amyloid precursor protein (APP), which has a role in iron efflux. This is via a classic release of repressor interaction of APP mRNA with iron-regulatory protein-1 (IRP1) whereas IRP2 controls the mRNAs encoding the L- and H-subunits of the iron storage protein, ferritin. Here, we identified thirteen potent APP translation blockers that acted selectively towards the uniquely configured iron-responsive element (IRE) RNA stem loop in the 5' untranslated region (UTR) of APP mRNA. These agents were 10-fold less inhibitory of 5'UTR sequences of the related prion protein (PrP) mRNA. Western blotting confirmed that the 'ninth' small molecule in the series selectively reduced neural APP production in SH-SY5Y cells at picomolar concentrations without affecting viability or the expression of α-synuclein and ferritin. APP blocker-9 (JTR-009), a benzimidazole, reduced the production of toxic Aβ in SH-SY5Y neuronal cells to a greater extent than other well tolerated APP 5'UTR-directed translation blockers, including posiphen, that were shown to limit amyloid burden in mouse models of Alzheimer's disease (AD). RNA binding assays demonstrated that JTR-009 operated by preventing IRP1 from binding to the IRE in APP mRNA, while maintaining IRP1 interaction with the H-ferritin IRE RNA stem loop. Thus, JTR-009 constitutively repressed translation driven by APP 5'UTR sequences. Calcein staining showed that JTR-009 did not indirectly change iron uptake in neuronal cells suggesting a direct interaction with the APP 5'UTR. These studies provide key data to develop small molecules that selectively reduce neural APP and Aβ production at 10-fold lower concentrations than related previously characterized translation blockers. Our data evidenced a novel therapeutic strategy of potential impact for people with trisomy of the APP gene on chromosome 21, which is a phenotype long associated with Down syndrome (DS) that can also cause familial Alzheimer's disease.
Collapse
Affiliation(s)
- Sanghamitra Bandyopadhyay
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, United States of America
| | - Catherine Cahill
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, United States of America
- Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, United States of America
| | - Amelie Balleidier
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, United States of America
| | - Conan Huang
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, United States of America
| | - Debomoy K. Lahiri
- Laboratory of Molecular Neurogenetics, Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Xudong Huang
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, United States of America
| | - Jack T. Rogers
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
25
|
Webster SJ, Bachstetter AD, Van Eldik LJ. Comprehensive behavioral characterization of an APP/PS-1 double knock-in mouse model of Alzheimer's disease. Alzheimers Res Ther 2013; 5:28. [PMID: 23705774 PMCID: PMC3706792 DOI: 10.1186/alzrt182] [Citation(s) in RCA: 111] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 04/29/2013] [Accepted: 05/24/2013] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Despite the extensive mechanistic and pathological characterization of the amyloid precursor protein (APP)/presenilin-1 (PS-1) knock-in mouse model of Alzheimer's disease (AD), very little is known about the AD-relevant behavioral deficits in this model. Characterization of the baseline behavioral performance in a variety of functional tasks and identification of the temporal onset of behavioral impairments are important to provide a foundation for future preclinical testing of AD therapeutics. Here we perform a comprehensive behavioral characterization of this model, discuss how the observed behavior correlates with the mechanistic and pathological observations of others, and compare this model with other commonly used AD mouse models. METHODS FOUR DIFFERENT GROUPS OF MICE RANGING ACROSS THE LIFESPAN OF THIS MODEL (TEST GROUPS: 7, 11, 15, and 24 months old) were run in a behavioral test battery consisting of tasks to assess motor function (grip strength, rotor rod, beam walk, open field ambulatory movement), anxiety-related behavior (open field time spent in peripheral zone vs. center zone, elevated plus maze), and cognitive function (novel object recognition, radial arm water maze). RESULTS There were no differences in motor function or anxiety-related behavior between APP/PS-1 knock-in mice and wild-type counterpart mice for any age group. Cognitive deficits in both recognition memory (novel object recognition) and spatial reference memory (radial arm water maze) became apparent for the knock-in animals as the disease progressed. CONCLUSION This is the first reported comprehensive behavioral analysis of the APP/PS1 knock-in mouse model of AD. The lack of motor/coordination deficits or abnormal anxiety levels, coupled with the age/disease-related cognitive decline and high physiological relevance of this model, make it well suited for utilization in preclinical testing of AD-relevant therapeutics.
Collapse
Affiliation(s)
- Scott J Webster
- Sanders-Brown Center on Aging, 800 S. Limestone, University of Kentucky, Lexington, KY 40536, USA
| | - Adam D Bachstetter
- Sanders-Brown Center on Aging, 800 S. Limestone, University of Kentucky, Lexington, KY 40536, USA
| | - Linda J Van Eldik
- Sanders-Brown Center on Aging, 800 S. Limestone, University of Kentucky, Lexington, KY 40536, USA
- Department of Anatomy and Neurobiology, 800 S. Limestone, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
26
|
Guan F, Gu J, Hu F, Zhu Y, Wang W. Association between α1-antichymotrypsin signal peptide -15A/T polymorphism and the risk of Alzheimer's disease: a meta-analysis. Mol Biol Rep 2012; 39:6661-9. [PMID: 22294107 DOI: 10.1007/s11033-012-1472-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2011] [Accepted: 01/23/2012] [Indexed: 11/26/2022]
Abstract
No consensus has been recently reached at the relationship between the α1-antichymotrypsin (ACT) signal peptide -15A/T polymorphism and Alzheimer’s disease (AD) risk. Thus, our study aimed to better assess this association by performing a meta-analysis, including 4,212 cases and 4,039 controls from 29 studies. Odds ratios (ORs) with the 95% confidence interval (CI) were used to assess the strength of relationship between ACT -15A/T polymorphism and AD risk. Overall, a borderline statistically significant association was detected under recessive model comparison in all subjects (AA vs. AT+TT: OR 1.12, 95% CI 1.01-1.25, P = 0.04). But in subgroup analysis by ethnicity, no significant association was found in Caucasians, Asians, or Africans. Moreover, after exclusion of one study which affect the heterogeneity, the ACT A allele and AA genotype were statistically associated with late-onset AD (LOAD) risk (AA vs. TT: OR 1.25, 95% CI 1.06-1.48, P = 0.007, A vs. T: OR 1.12, 95% CI 1.03-1.21, P = 0.008), especially in Caucasians. In conclusion, our study suggests that the common α1-antichymotrypsin signal peptide -15A/T polymorphism may not be a major risk factor for AD. However, the polymorphism is capable of increasing LOAD risk.
Collapse
Affiliation(s)
- Fulin Guan
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | | | | | | | | |
Collapse
|
27
|
Nilsson LNG, Gografe S, Costa DA, Hughes T, Dressler D, Potter H. USE OF FUSED CIRCULATIONS TO INVESTIGATE THE ROLE OF APOLIPOPROTEIN E AS AMYLOID CATALYST AND PERIPHERAL SINK IN ALZHEIMER'S DISEASE. TECHNOLOGY AND INNOVATION 2012; 14:199-208. [PMID: 23626867 DOI: 10.3727/194982412x13462021398010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Apolipoprotein E (apoE) synthesized in liver and brain plays a key role in both cholesterol transport and Alzheimer's disease (AD): apoE-knockout mice develop hypercholesterolemia and atherosclerosis and cannot support AD amyloid deposition. The ApoE4 allele is the strongest genetic risk factor for late-onset AD, and apoE4 protein preferentially catalyzes amyloid-beta (Aβ) peptide fibrillization in vitro and amyloid plaque deposition in vivo. Circulating apoE may also have the potential to draw Aβ from the brain and reduce amyloid deposition. We used parabiosis to determine how circulating apoE impacts brain amyloid deposition and blood cholesterol levels in transgenic mice carrying AD-promoting APP and PS1 human transgenes-either with or without the endogenous mouse apoE gene. ApoE transferred through the joined circulations from WT to parabiosed APP+/+,PS1+/-,apoE-KO mice prevented hypercholesterolemia and reduced already low brain amyloid deposition. The findings indicate that apoE synthesis in the brain itself is necessary for amyloid accumulation. Furthermore, plasma apoE can both normalize cholesterol levels in apoE-KO mice and act as a peripheral sink to induce net efflux of Aβ peptide from the brain. The therapeutic implication is that inhibiting Alzheimer's disease neuropathology may be accomplished by either reducing apoE in the brain or increasing apoE in the blood.
Collapse
Affiliation(s)
- Lars N G Nilsson
- Department of Molecular Medicine and Suncoast Gerontology Center, University of South Florida, Tampa, FL, USA ; Department of Pharmacology, Faculty of Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | | | | | | | | | | |
Collapse
|
28
|
Lord A, Philipson O, Klingstedt T, Westermark G, Hammarström P, Nilsson KPR, Nilsson LNG. Observations in APP bitransgenic mice suggest that diffuse and compact plaques form via independent processes in Alzheimer's disease. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 178:2286-98. [PMID: 21514441 DOI: 10.1016/j.ajpath.2011.01.052] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2010] [Revised: 01/04/2011] [Accepted: 01/13/2011] [Indexed: 11/26/2022]
Abstract
Studies of familial Alzheimer's disease suggest that misfolding and aggregation of amyloid-β (Aβ) peptides initiate the pathogenesis. The Arctic mutation of Aβ precursor protein (APP) results in AD, and Arctic Aβ is more prone to form Aβ protofibrils and extracellular deposits. Herein is demonstrated that the burden of diffuse Aβ deposits but not compact plaques is increased when tg-Swe mice are crossed with tg-ArcSwe mice synthesizing low levels of Arctic Aβ. The diffuse deposits in bitransgenic mice, which contain primarily wild-type Aβ42, accumulate in regions both with and without transgene expression. However, APP processing, when compared with tg-Swe, remains unchanged in young bitransgenic mice, whereas wild-type Aβ42 aggregation is accelerated and fibril architecture is altered in vitro and in vivo when a low level of Arctic Aβ42 is introduced. Thus, the increased number of diffuse deposits is likely due to physical interactions between Arctic Aβ and wild-type Aβ42. The selective increase of a single type of parenchymal Aβ deposit suggests that different pathways lead to formation of diffuse and compact plaques. These findings could have general implications for Alzheimer's disease pathogenesis and particular relevance to patients heterozygous for the Arctic APP mutation. Moreover, it further illustrates how Aβ neuropathologic features can be manipulated in vivo by mechanisms similar to those originally conceptualized in prion research.
Collapse
Affiliation(s)
- Anna Lord
- Department of Public Health and Caring Sciences/Molecular Geriatrics, Uppsala University, Uppsala, Sweden
| | | | | | | | | | | | | |
Collapse
|
29
|
Roles of apolipoprotein E4 (ApoE4) in the pathogenesis of Alzheimer's disease: lessons from ApoE mouse models. Biochem Soc Trans 2011; 39:924-32. [DOI: 10.1042/bst0390924] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
ApoE4 (apolipoprotein E4) is the major known genetic risk factor for AD (Alzheimer's disease). In most clinical studies, apoE4 carriers account for 65–80% of all AD cases, highlighting the importance of apoE4 in AD pathogenesis. Emerging data suggest that apoE4, with its multiple cellular origins and multiple structural and biophysical properties, contributes to AD in multiple ways either independently or in combination with other factors, such as Aβ (amyloid β-peptide) and tau. Many apoE mouse models have been established to study the mechanisms underlying the pathogenic actions of apoE4. These include transgenic mice expressing different apoE isoforms in neurons or astrocytes, those expressing neurotoxic apoE4 fragments in neurons and human apoE isoform knock-in mice. Since apoE is expressed in different types of cells, including astrocytes and neurons, and in brains under diverse physiological and/or pathophysiological conditions, these apoE mouse models provide unique tools to study the cellular source-dependent roles of apoE isoforms in neurobiology and in the pathogenesis of AD. They also provide useful tools for discovery and development of drugs targeting apoE4's detrimental effects.
Collapse
|
30
|
Boyd TD, Bennett SP, Mori T, Governatori N, Runfeldt M, Norden M, Padmanabhan J, Neame P, Wefes I, Sanchez-Ramos J, Arendash GW, Potter H. GM-CSF upregulated in rheumatoid arthritis reverses cognitive impairment and amyloidosis in Alzheimer mice. J Alzheimers Dis 2010; 21:507-18. [PMID: 20555144 DOI: 10.3233/jad-2010-091471] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Rheumatoid arthritis (RA) is a negative risk factor for the development of Alzheimer's disease (AD). While it has been commonly assumed that RA patients' usage of non-steroidal anti-inflammatory drugs (NSAIDs) helped prevent onset and progression of AD, NSAID clinical trials have proven unsuccessful in AD patients. To determine whether intrinsic factors within RA pathogenesis itself may underlie RA's protective effect, we investigated the activity of colony-stimulating factors, upregulated in RA, on the pathology and behavior of transgenic AD mice. 5 microg bolus injections of macrophage, granulocyte, and granulocyte-macrophage colony-stimulating factors (M-CSF, G-CSF, or GM-CSF) were administered unilaterally into the hippocampus of aged cognitively-impaired AD mice and the resulting amyloid load reductions determined one week later, using the artificial cerebrospinal fluid-injected contralateral sides as controls. G-CSF and more significantly, GM-CSF reduced amyloidosis throughout the treated brain hemisphere one week following bolus administration to AD mice. 20 daily subcutaneous injections of 5 microg of GM-CSF (the most amyloid-reducing CSF in the bolus experiment) were administered to balanced cohorts of AD mice after assessment in a battery of cognitive tests. Reductions in amyloid load and improvements in cognitive function were assessed. Subcutaneous GM-CSF administration significantly reduced brain amyloidosis and completely reversed the cognitive impairment, while increasing hippocampal synaptic area and microglial density. These findings, along with two decades of accrued safety data using Leukine, recombinant human GMCSF, in elderly leukopenic patients, suggest that Leukine should be tested as a treatment to reverse cerebral amyloid pathology and cognitive impairment in AD.
Collapse
Affiliation(s)
- Tim D Boyd
- USF Health Byrd Alzheimer's Center and Research Institute, Tampa, FL 33613, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Ianni M, Manerba M, Di Stefano G, Porcellini E, Chiappelli M, Carbone I, Licastro F. Altered glycosylation profile of purified plasma ACT from Alzheimer's disease. Immun Ageing 2010; 7 Suppl 1:S6. [PMID: 21172065 PMCID: PMC3024880 DOI: 10.1186/1742-4933-7-s1-s6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is one of the most frequent cause of neurodegenerative disorder in the elderly. Inflammation has been implicated in brain degenerative processes and peripheral markers of brain AD related impairment would be useful. Plasma levels of alpha-1-antichymotrypsin (ACT), an acute phase protein and a secondary component of amyloid plaques, are often increased in AD patients and high blood ACT levels correlate with progressive cognitive deterioration. During inflammatory responses changes in the micro-heterogeneity of ACT sugar chains have been described. METHODS N-Glycanase digestion from Flavobacterium meningosepticum (PNGase F) was performed on both native and denatured purified ACT condition and resolved to Western blot with the purpose to revealed the ACT de-glycosylation pattern.Further characterization of the ACT glycan profile was obtained by a glycoarray; each lectin group in the assay specifically recognizes one or two glycans/epitopes. Lectin-bound ACT produced a glyco-fingerprint and mayor differences between AD and controls samples were assessed by a specific algorithms. RESULTS Western blot analysis of purified ACT after PNGase F treatment and analysis of sugar composition of ACT showed significantly difference in "glyco-fingerprints" patterns from controls (CTR) and AD; ACT from AD showing significantly reduced levels of sialic acid. A difference in terminal GlcNac residues appeared to be related with progressive cognitive deterioration. CONCLUSIONS Low content of terminal GlcNac and sialic acid in peripheral ACT in AD patients suggests that a different pattern of glycosylation might be a marker of brain inflammation. Moreover ACT glycosylation analysis could be used to predict AD clinical progression and used in clinical trials as surrogate marker of clinical efficacy.
Collapse
Affiliation(s)
- Manuela Ianni
- Department of Experimental Pathology, School of Medicine, University of Bologna, 14 San Giacomo St, 40126 Bologna, Italy
| | - Marcella Manerba
- Department of Experimental Pathology, School of Medicine, University of Bologna, 14 San Giacomo St, 40126 Bologna, Italy
| | - Giuseppina Di Stefano
- Department of Experimental Pathology, School of Medicine, University of Bologna, 14 San Giacomo St, 40126 Bologna, Italy
| | - Elisa Porcellini
- Department of Experimental Pathology, School of Medicine, University of Bologna, 14 San Giacomo St, 40126 Bologna, Italy
| | - Martina Chiappelli
- Department of Experimental Pathology, School of Medicine, University of Bologna, 14 San Giacomo St, 40126 Bologna, Italy
| | - Ilaria Carbone
- Department of Experimental Pathology, School of Medicine, University of Bologna, 14 San Giacomo St, 40126 Bologna, Italy
| | - Federico Licastro
- Department of Experimental Pathology, School of Medicine, University of Bologna, 14 San Giacomo St, 40126 Bologna, Italy
| |
Collapse
|
32
|
Vasilevko V, Passos G, Quiring D, Head E, Fisher M, Cribbs DH. Aging and cerebrovascular dysfunction: contribution of hypertension, cerebral amyloid angiopathy, and immunotherapy. Ann N Y Acad Sci 2010; 1207:58-70. [PMID: 20955427 PMCID: PMC2958685 DOI: 10.1111/j.1749-6632.2010.05786.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Age-related cerebrovascular dysfunction contributes to ischemic stroke, intracerebral hemorrhages (ICHs), microbleeds, cerebral amyloid angiopathy (CAA), and cognitive decline. Importantly, there is increasing recognition that this dysfunction plays a critical secondary role in many neurodegenerative diseases, including Alzheimer's disease (AD). Atherosclerosis, hypertension, and CAA are the most common causes of blood-brain barrier (BBB) lesions. The accumulation of amyloid beta (Aβ) in the cerebrovascular system is a significant risk factor for ICH and has been linked to endothelial transport failure and blockage of perivascular drainage. Moreover, recent anti-Aβ immunotherapy clinical trials demonstrated efficient clearance of parenchymal amyloid deposits but have been plagued by CAA-associated adverse events. Although management of hypertension and atherosclerosis can reduce the incidence of ICH, there are currently no approved therapies for attenuating CAA. Thus, there is a critical need for new strategies that improve BBB function and limit the development of β-amyloidosis in the cerebral vasculature.
Collapse
Affiliation(s)
- Vitaly Vasilevko
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA
| | - Giselle Passos
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA
| | - Daniel Quiring
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA
| | - Elizabeth Head
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA
- Department of Neurology, University of California, Irvine, Irvine, CA
| | - Mark Fisher
- Department of Neurology, University of California, Irvine, Irvine, CA
| | - David H. Cribbs
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA
- Department of Neurology, University of California, Irvine, Irvine, CA
| |
Collapse
|
33
|
Abstract
PURPOSE OF REVIEW The purpose of this review is to provide insights into recent advances in mechanisms linking apolipoprotein (apo) E isoforms to cardiovascular and neurological diseases. RECENT FINDINGS Human apoE has three common isoforms (apoE2, apoE3, and apoE4) with different structural and biophysical properties and different effects on lipid and neuronal homeostasis. ApoE is a protein constituent of different plasma lipoproteins and serves as a high-affinity ligand for several receptors. By interacting with its receptors, apoE mediates the clearance of different lipoproteins from the circulation. Absence or structural mutations of apoE cause significant disorders in lipid metabolism and cardiovascular disease. ApoE also has significant roles in neurobiology. ApoE4 is the major known genetic risk factor for Alzheimer's disease. It increases the occurrence and lowers the age of onset of Alzheimer's disease. ApoE4 carriers account for 65-80% of all Alzheimer's disease cases, highlighting the importance of apoE4 in Alzheimer's disease pathogenesis. ApoE4 has both amyloid beta-dependent and amyloid beta-independent roles in Alzheimer's disease pathogenesis. SUMMARY Emerging data suggest that apoE isoforms, with their multiple cellular origins and multiple structural and biophysical properties, contribute to cardiovascular and neurological diseases by interacting with different factors through various pathways.
Collapse
Affiliation(s)
- Yadong Huang
- Gladstone Institute of Neurological Disease, University of California, San Francisco, California 94158, USA.
| |
Collapse
|
34
|
Imbimbo BP, Solfrizzi V, Panza F. Are NSAIDs useful to treat Alzheimer's disease or mild cognitive impairment? Front Aging Neurosci 2010; 2. [PMID: 20725517 PMCID: PMC2912027 DOI: 10.3389/fnagi.2010.00019] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2010] [Accepted: 04/23/2010] [Indexed: 11/13/2022] Open
Abstract
Several epidemiological studies suggest that long-term use of non-steroidal anti-inflammatory drugs (NSAIDs) may protect subjects carrying one or more ε4 allele of the apolipoprotein E (APOE ε4) against the onset of Alzheimer's disease (AD). The biological mechanism of this protection is not completely understood and may involve the anti-inflammatory properties of NSAIDs or their ability of interfering with the β-amyloid (Aβ) cascade. Unfortunately, long-term, placebo-controlled clinical trials with both non-selective and cyclooxygenase-2 (COX-2) selective inhibitors in mild-to-moderate AD patients produced negative results. A secondary prevention study with rofecoxib, a COX-2 selective inhibitor, in patients with mild cognitive impairment was also negative. A primary prevention study (ADAPT trial) of naproxen (a non-selective COX inhibitor) and celecoxib (a COX-2 selective inhibitor) in cognitively normal elderly subjects with a family history of AD was prematurely interrupted for safety reasons after a median period of treatment of 2 years. Although both drugs did not reduce the incidence of dementia after 2 years of treatment, a 4-year follow-up assessment surprisingly revealed that subjects previously exposed to naproxen were protected from the onset of AD by 67% compared to placebo. Thus, it could be hypothesized that the chronic use of NSAIDs may be beneficial only in the very early stages of the AD process in coincidence of initial Aβ deposition, microglia activation and consequent release of pro-inflammatory mediators. When the Aβ deposition process is already started, NSAIDs are no longer effective and may even be detrimental because of their inhibitory activity on chronically activated microglia that on long-term may mediate Aβ clearance. The research community should conduct long-term trials with NSAIDs in cognitively normal APOE ε4 carriers.
Collapse
Affiliation(s)
- Bruno P Imbimbo
- Research and Development Department, Chiesi Farmaceutici Parma, Italy
| | | | | |
Collapse
|
35
|
Philipson O, Lord A, Gumucio A, O'Callaghan P, Lannfelt L, Nilsson LNG. Animal models of amyloid-beta-related pathologies in Alzheimer's disease. FEBS J 2010; 277:1389-409. [PMID: 20136653 DOI: 10.1111/j.1742-4658.2010.07564.x] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
In the early 1990s, breakthrough discoveries on the genetics of Alzheimer's disease led to the identification of missense mutations in the amyloid-beta precursor protein gene. Research findings quickly followed, giving insights into molecular pathogenesis and possibilities for the development of new types of animal models. The complete toolbox of transgenic techniques, including pronuclear oocyte injection and homologous recombination, has been applied in the Alzheimer's disease field, to produce overexpressors, knockouts, knockins and regulatable transgenics. Transgenic models have dramatically advanced our understanding of pathogenic mechanisms and allowed therapeutic approaches to be tested. Following a brief introduction to Alzheimer's disease, various nontransgenic and transgenic animal models are described in terms of their values and limitations with respect to pathogenic, therapeutic and functional understandings of the human disease.
Collapse
Affiliation(s)
- Ola Philipson
- Department of Public Health and Caring Sciences/Molecular Geriatrics, Uppsala University, Sweden
| | | | | | | | | | | |
Collapse
|
36
|
Bruinsma IB, Wilhelmus MMM, Kox M, Veerhuis R, de Waal RMW, Verbeek MM. Apolipoprotein E protects cultured pericytes and astrocytes from D-Abeta(1-40)-mediated cell death. Brain Res 2009; 1315:169-80. [PMID: 20034483 DOI: 10.1016/j.brainres.2009.12.039] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2009] [Revised: 12/10/2009] [Accepted: 12/12/2009] [Indexed: 12/15/2022]
Abstract
Cerebral amyloid angiopathy (CAA) is a common pathological finding in Alzheimer's disease and hereditary cerebral hemorrhage with amyloidosis of the Dutch type; in this latter condition it is caused by deposition of mutated amyloid beta protein (Abeta Glu22Gln; D-Abeta(1-40)). Previously, we found a dependence of the Abeta-mediated toxicity and apolipoprotein E (apoE) production by cultured pericytes on apoE genotype. Given their close association with the cerebrovascular wall both astrocytes and pericytes may be involved in CAA development, a process that includes Abeta deposition and clearance and that may be affected by interaction with locally produced apolipoprotein E (apoE). Although astrocytes are regarded as the major source of apolipoprotein E (apoE) in the brain, also pericytes produce apoE. In this study we compared the apoE production capacity, the effects of apoE on D-Abeta(1-40) internalization, D-Abeta(1-40) cell surface accumulation and the vulnerability for D-Abeta(1-40)-induced toxicity of either cell type in order to quantify the relative contributions of astrocytes and pericytes in the various processes that contribute to CAA formation. Strikingly, cultured astrocytes produced only 3-10% of the apoE amounts produced by pericytes. Furthermore, pericytes with the apoE epsilon4 allele produced three times less apoE and were more vulnerable to D-Abeta(1-40) treatment than pericytes without an epsilon4 allele. Such relations were not observed with astrocytes in vitro. Both pericytes and astrocytes, however, were protected from Abeta-induced cytotoxicity by high levels of pericyte-derived apoE, but not recombinant apoE. In addition, pericyte-derived apoE dose-dependently decreased both internalization of Abeta and Abeta accumulation at the cell surface in either cell type. The present data suggest that apoE produced by pericytes, rather than astrocyte-produced apoE, modulates Abeta cytotoxicity and Abeta removal near the vasculature in the brain. Furthermore, since apoE production in pericytes is genotype dependent, this may contribute to the apoE genotype-dependent development of CAA in vivo.
Collapse
Affiliation(s)
- Ilona B Bruinsma
- Radboud University Nijmegen Medical Centre, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands.
| | | | | | | | | | | |
Collapse
|
37
|
Olcese JM, Cao C, Mori T, Mamcarz MB, Maxwell A, Runfeldt MJ, Wang L, Zhang C, Lin X, Zhang G, Arendash GW. Protection against cognitive deficits and markers of neurodegeneration by long-term oral administration of melatonin in a transgenic model of Alzheimer disease. J Pineal Res 2009; 47:82-96. [PMID: 19538338 DOI: 10.1111/j.1600-079x.2009.00692.x] [Citation(s) in RCA: 172] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The neurohormone melatonin has been reported to exert anti-beta-amyloid aggregation, antioxidant, and anti-inflammatory actions in various in vitro and animal models. To comprehensively determine the potential for long-term melatonin treatment to protect Alzheimer's transgenic mice against cognitive impairment and development of beta-amyloid (Abeta) neuropathology, we administered melatonin (100 mg/L drinking water) to APP + PS1 double transgenic (Tg) mice from 2-2.5 months of age to their killing at age 7.5 months. A comprehensive behavioral battery administered during the final 6 weeks of treatment revealed that Tg mice given melatonin were protected from cognitive impairment in a variety of tasks of working memory, spatial reference learning/memory, and basic mnemonic function; Tg control mice remained impaired in all of these cognitive tasks/domains. Immunoreactive Abeta deposition was significantly reduced in hippocampus (43%) and entorhinal cortex (37%) of melatonin-treated Tg mice. Although soluble and oligomeric forms of Abeta1-40 and 1-42 were unchanged in the hippocampus and cortex of the same melatonin-treated Tg mice, their plasma Abeta levels were elevated. These Abeta results, together with our concurrent demonstration that melatonin suppresses Abeta aggregation in brain homogenates, are consistent with a melatonin-facilitated removal of Abeta from the brain. Inflammatory cytokines such as tumor necrosis factor (TNF)-alpha were decreased in hippocampus (but not plasma) of Tg+ melatonin mice. Finally, the cortical mRNA expression of three antioxidant enzymes (SOD-1, glutathione peroxidase, and catalase) was significantly reduced to non-Tg levels by long-term melatonin treatment in Tg mice. Thus, melatonin's cognitive benefits could involve its anti-Abeta aggregation, anti-inflammatory, and/or antioxidant properties. Our findings provide support for long-term melatonin therapy as a primary or complementary strategy for abating the progression of Alzheimer disease.
Collapse
Affiliation(s)
- James M Olcese
- Florida State University College of Medicine, Tallahassee, FL, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Lord A, Englund H, Söderberg L, Tucker S, Clausen F, Hillered L, Gordon M, Morgan D, Lannfelt L, Pettersson FE, Nilsson LNG. Amyloid-beta protofibril levels correlate with spatial learning in Arctic Alzheimer's disease transgenic mice. FEBS J 2009; 276:995-1006. [PMID: 19215301 DOI: 10.1111/j.1742-4658.2008.06836.x] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Oligomeric assemblies of amyloid-beta (Abeta) are suggested to be central in the pathogenesis of Alzheimer's disease because levels of soluble Abeta correlate much better with the extent of cognitive dysfunctions than do senile plaque counts. Moreover, such Abeta species have been shown to be neurotoxic, to interfere with learned behavior and to inhibit the maintenance of hippocampal long-term potentiation. The tg-ArcSwe model (i.e. transgenic mice with the Arctic and Swedish Alzheimer mutations) expresses elevated levels of Abeta protofibrils in the brain, making tg-ArcSwe a highly suitable model for investigating the pathogenic role of these Abeta assemblies. In the present study, we estimated Abeta protofibril levels in the brain and cerebrospinal fluid of tg-ArcSwe mice, and also assessed their role with respect to cognitive functions. Protofibril levels, specifically measured with a sandwich ELISA, were found to be elevated in young tg-ArcSwe mice compared to several transgenic models lacking the Arctic mutation. In aged tg-ArcSwe mice with considerable plaque deposition, Abeta protofibrils were approximately 50% higher than in younger mice, whereas levels of total Abeta were exponentially increased. Young tg-ArcSwe mice showed deficits in spatial learning, and individual performances in the Morris water maze were correlated inversely with levels of Abeta protofibrils, but not with total Abeta levels. We conclude that Abeta protofibrils accumulate in an age-dependent manner in tg-ArcSwe mice, although to a far lesser extent than total Abeta. Our findings suggest that increased levels of Abeta protofibrils could result in spatial learning impairment.
Collapse
Affiliation(s)
- Anna Lord
- Department of Public Health and Caring Sciences/Molecular Geriatrics, Uppsala University, Sweden
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Mutant Amyloid-beta-sensitized dendritic cells as Alzheimer's disease vaccine. J Neuroimmunol 2008; 200:1-10. [DOI: 10.1016/j.jneuroim.2008.05.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2008] [Revised: 05/20/2008] [Accepted: 05/20/2008] [Indexed: 12/29/2022]
|
40
|
Belbin O, Dunn JL, Chappell S, Ritchie AE, Ling Y, Morgan L, Pritchard A, Warden DR, Lendon CL, Lehmann DJ, Mann DMA, Smith AD, Kalsheker N, Morgan K. A SNP in the ACT gene associated with astrocytosis and rapid cognitive decline in AD. Neurobiol Aging 2008; 29:1167-76. [PMID: 17368652 DOI: 10.1016/j.neurobiolaging.2007.02.021] [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: 11/15/2006] [Revised: 12/19/2006] [Accepted: 02/10/2007] [Indexed: 11/27/2022]
Abstract
There is biochemical and animal model evidence supporting a pathological role of the ACT gene in AD. However, direct genetic evidence remains controversial and has been mostly limited to individual single nucleotide polymorphism (SNP) analysis. To resolve this apparent conflict we have used a high-density ACT SNP map, constructed haplotypes and explored correlations with phenotype. SNPs were identified by sequencing and used to construct haplotypes in 668 AD patients and 419 controls and a case-control association study was performed. Five SNPs, comprising five common haplotypes, represented 93% of ACT gene variation. Although no single SNP or haplotype was associated with AD status, a SNP in intron 2 was associated with later onset and more rapid cognitive decline (P=0.04). This SNP was both individually associated with severe astrocytosis (P=0.004) in AD patients and when combined with the signal sequence SNP (P=0.002). This suggests that astrocytosis may have a protective function for a limited period in some patients. These SNP associations either support a direct role for the ACT gene, in AD pathology or alternatively reflect linkage with polymorphisms in other genes nearby.
Collapse
Affiliation(s)
- O Belbin
- Division of Clinical Chemistry, Institute of Genetics, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Cedazo-Mínguez A. Apolipoprotein E and Alzheimer's disease: molecular mechanisms and therapeutic opportunities. J Cell Mol Med 2008; 11:1227-38. [PMID: 18205697 PMCID: PMC4401287 DOI: 10.1111/j.1582-4934.2007.00130.x] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Multiple genetic and environmental factors are likely to contribute to the development of Alzheimer's disease (AD). The most important known risk factor for AD is presence of the E4 isoform of apolipoprotein E (apoE). Epidemiological studies demonstrated that apoE4 carriers have a higher risk and develop the disease and an early onset. Moreover, apoE4 is the only molecule that has been associated with all the biochemical disturbances characteristic of the disease: amyloid-beta (Abeta) deposition, tangle formation, oxidative stress, lipid homeostasis deregulation, synaptic plasticity loss and cholinergic dysfunction. This large body of evidence suggest that apoE is a key player in the pathogenesis of AD. This short review examines the current facts and hypotheses of the association between apoE4 and AD, as well as the therapeutic possibilities that apoE might offer for the treatment of this disease.
Collapse
Affiliation(s)
- Angel Cedazo-Mínguez
- Karolinska Institutet, Department of Neurobiology, Care Sciences and Society, KI-Alzheimer's Disease Research Center, NOVUM, Stockholm, Sweden.
| |
Collapse
|
42
|
Cao C, Lin X, Wahi MM, Jackson EA, Potter H. Successful adjuvant-free vaccination of BALB/c mice with mutated amyloid beta peptides. BMC Neurosci 2008; 9:25. [PMID: 18282292 PMCID: PMC2270279 DOI: 10.1186/1471-2202-9-25] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2006] [Accepted: 02/18/2008] [Indexed: 02/05/2023] Open
Abstract
Background A recent human clinical trial of an Alzheimer's disease (AD) vaccine using amyloid beta (Aβ) 1–42 plus QS-21 adjuvant produced some positive results, but was halted due to meningoencephalitis in some participants. The development of a vaccine with mutant Aβ peptides that avoids the use of an adjuvant may result in an effective and safer human vaccine. Results All peptides tested showed high antibody responses, were long-lasting, and demonstrated good memory response. Epitope mapping indicated that peptide mutation did not lead to epitope switching. Mutant peptides induced different inflammation responses as evidenced by cytokine profiles. Ig isotyping indicated that adjuvant-free vaccination with peptides drove an adequate Th2 response. All anti-sera from vaccinated mice cross-reacted with human Aβ in APP/PS1 transgenic mouse brain tissue. Conclusion Our study demonstrated that an adjuvant-free vaccine with different Aβ peptides can be an effective and safe vaccination approach against AD. This study represents the first report of adjuvant-free vaccines utilizing Aβ peptides carrying diverse mutations in the T-cell epitope. These largely positive results provide encouragement for the future of the development of human vaccinations for AD.
Collapse
Affiliation(s)
- Chuanhai Cao
- Johnnie B, Byrd Alzheimer's Center and Research Institute, 4001 E, Fletcher Ave,, Third Floor, Tampa, FL 33613, USA.
| | | | | | | | | |
Collapse
|
43
|
Arendash GW, Jensen MT, Salem N, Hussein N, Cracchiolo J, Dickson A, Leighty R, Potter H. A diet high in omega-3 fatty acids does not improve or protect cognitive performance in Alzheimer’s transgenic mice. Neuroscience 2007; 149:286-302. [PMID: 17904756 DOI: 10.1016/j.neuroscience.2007.08.018] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2007] [Revised: 07/17/2007] [Accepted: 08/04/2007] [Indexed: 10/23/2022]
Abstract
Although a number of epidemiologic studies reported that higher intake of omega-3 fatty acids (largely associated with fish consumption) is protective against Alzheimer's disease (AD), other human studies reported no such effect. Because retrospective human studies are problematic and controlled longitudinal studies over decades are impractical, the present study utilized Alzheimer's transgenic mice (Tg) in a highly controlled study to determine whether a diet high in omega-3 fatty acid, equivalent to the 13% omega-3 fatty acid diet of Greenland Eskimos, can improve cognitive performance or protect against cognitive impairment. Amyloid precursor protein (APP)-sw+PS1 double transgenic mice, as well as nontransgenic (NT) normal littermates, were given a high omega-3 supplemented diet or a standard diet from 2 through 9 months of age, with a comprehensive behavioral test battery administered during the final 6 weeks. For both Tg and NT mice, long-term n-3 supplementation resulted in cognitive performance that was no better than that of mice fed a standard diet. In NT mice, the high omega-3 diet increased cortical levels of omega-3 fatty acids while decreasing omega-6 levels. However, the high omega-3 diet had no effect on cortical fatty acid levels in Tg mice. Irrespective of diet, no correlations existed between brain omega-3 levels and cognitive performance for individual NT or Tg mice. In contrast, brain levels of omega-6 fatty acids were strongly correlated with cognitive impairment for both genotypes. Thus, elevated brain levels of omega-3 fatty acids were not relevant to cognitive function, whereas high brain levels of omega-6 were associated with impaired cognitive function. In Tg mice, the omega-3 supplemental diet did not induce significant changes in soluble/insoluble Abeta within the hippocampus, although strong correlations were evident between hippocampal Abeta(1-40) levels and cognitive impairment. While these studies involved a genetically manipulated mouse model of AD, our results suggest that diets high in omega-3 fatty acids, or use of fish oil supplements (DHA+EPA), will not protect against AD, at least in high-risk individuals. However, normal individuals conceivably could derive cognitive benefits from high omega-3 intake if it corrects an elevation in the brain level of n-6 fatty acids as a result. Alternatively, dietary fish may contain nutrients, other than DHA and EPA, that could provide some protection against AD.
Collapse
Affiliation(s)
- G W Arendash
- The Florida Alzheimer's Disease Research Center and The Byrd Alzheimer's Institute, Tampa, FL 33613, USA.
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Cracchiolo JR, Mori T, Nazian SJ, Tan J, Potter H, Arendash GW. Enhanced cognitive activity--over and above social or physical activity--is required to protect Alzheimer's mice against cognitive impairment, reduce Abeta deposition, and increase synaptic immunoreactivity. Neurobiol Learn Mem 2007; 88:277-94. [PMID: 17714960 PMCID: PMC2083653 DOI: 10.1016/j.nlm.2007.07.007] [Citation(s) in RCA: 113] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2007] [Revised: 06/21/2007] [Accepted: 07/05/2007] [Indexed: 11/25/2022]
Abstract
Although social, physical, and cognitive activities have each been suggested to reduce the risk of Alzheimer's disease (AD), epidemiologic studies cannot determine which activity or combination of activities is most important. To address this question, mutant APP transgenic AD mice were reared long-term in one of four housing conditions (impoverished, social, social+physical, or complete enrichment) from 1(1/2) through 9 months of age. Thus, a stepwise layering of social, physical, and enhanced cognitive activity was created. Behavioral evaluation in a full battery of sensorimotor, anxiety, and cognitive tasks was carried out during the final 5 weeks of housing. Only AD mice raised in complete enrichment (i.e., enhanced cognitive activity) showed: (1) protection against cognitive impairment, (2) decreased brain beta-amyloid deposition, and (3) increased hippocampal synaptic immunoreactivity. The protection provided by enhanced cognitive activity spanned multiple cognitive domains (working memory, reference learning, and recognition/identification). Cognitive and neurohistologic benefits of complete enrichment occurred without any changes in blood cytokine or corticosterone levels, suggesting that enrichment-dependent mechanisms do not involve changes in the inflammatory response or stress levels, respectively. These results indicate that the enhanced cognitive activity of complete enrichment is required for cognitive and neurologic benefit to AD mice-physical and/or social activity are insufficient. Thus, our data suggest that humans who emphasize a high lifelong level of cognitive activity (over and above social and physical activities) will attain the maximal environmental protection against AD.
Collapse
Affiliation(s)
- Jennifer R. Cracchiolo
- The Johnnie B. Byrd Alzheimer’s Center and Research Institute, Tampa, FL 33613
- Florida Alzheimer’s Disease Research Center, University of South Florida, Tampa FL 33612
| | - Takashi Mori
- Institute of Medical Science, Department of Pathology, Saitama Medical Center/Saitama Medical University, Saitama 350-8550, Japan
| | - Stanley J. Nazian
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL 33612
| | - Jun Tan
- Department of Psychiatry and Behavioral Medicine, University of South Florida, Tampa, FL 33612
| | - Huntington Potter
- The Johnnie B. Byrd Alzheimer’s Center and Research Institute, Tampa, FL 33613
- Florida Alzheimer’s Disease Research Center, University of South Florida, Tampa FL 33612
| | - Gary W. Arendash
- The Johnnie B. Byrd Alzheimer’s Center and Research Institute, Tampa, FL 33613
- Florida Alzheimer’s Disease Research Center, University of South Florida, Tampa FL 33612
- *Correspondence to: Gary W. Arendash, Ph.D., The Byrd Alzheimer’s Center & Research Institute, Tampa, FL 33613, Phone: (813) 974-1584, FAX: (813) 866-1601, Email
| |
Collapse
|
45
|
Costa DA, Cracchiolo JR, Bachstetter AD, Hughes TF, Bales KR, Paul SM, Mervis RF, Arendash GW, Potter H. Enrichment improves cognition in AD mice by amyloid-related and unrelated mechanisms. Neurobiol Aging 2007; 28:831-44. [PMID: 16730391 DOI: 10.1016/j.neurobiolaging.2006.04.009] [Citation(s) in RCA: 170] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2005] [Revised: 03/31/2006] [Accepted: 04/07/2006] [Indexed: 11/27/2022]
Abstract
Lifelong cognitive stimulation is associated with a lower risk of Alzheimer's disease (AD), but causality is difficult to prove. We therefore sought to investigate the preventative potential of environmental enrichment (EE) using mice expressing both human mutant presenilin-1 and the amyloid precursor protein (PS1/PDAPP). At weaning, mice were placed into either an enriched or standard housing environment. Behavioral testing at 4.5-6 months showed that environmentally enriched PS1/PDAPP mice outperformed mice in standard housing, and were behaviorally indistinguishable from non-transgenic mice across multiple cognitive domains. PS1/PDAPP mice exposed to both environmental enrichment and behavioral testing, but not to EE alone, showed 50% less brain beta-amyloid without improved dendritic morphology. Microarray analysis revealed large enrichment-induced changes in hippocampal expression of genes/proteins related to Abeta sequestration and synaptic plasticity. These results indicate that EE protects against cognitive impairment in AD transgenic mice through a dual mechanism, including both amyloid dependent and independent mechanisms.
Collapse
Affiliation(s)
- David A Costa
- Johnnie B. Byrd Sr. Alzheimer's Center and Research Institute, Tampa, FL 33647, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Pathways and genes differentially expressed in the motor cortex of patients with sporadic amyotrophic lateral sclerosis. BMC Genomics 2007; 8:26. [PMID: 17244347 PMCID: PMC1796866 DOI: 10.1186/1471-2164-8-26] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2006] [Accepted: 01/23/2007] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is a fatal disorder caused by the progressive degeneration of motoneurons in brain and spinal cord. Despite identification of disease-linked mutations, the diversity of processes involved and the ambiguity of their relative importance in ALS pathogenesis still represent a major impediment to disease models as a basis for effective therapies. Moreover, the human motor cortex, although critical to ALS pathology and physiologically altered in most forms of the disease, has not been screened systematically for therapeutic targets. RESULTS By whole-genome expression profiling and stringent significance tests we identify genes and gene groups de-regulated in the motor cortex of patients with sporadic ALS, and interpret the role of individual candidate genes in a framework of differentially expressed pathways. Our findings emphasize the importance of defense responses and cytoskeletal, mitochondrial and proteasomal dysfunction, reflect reduced neuronal maintenance and vesicle trafficking, and implicate impaired ion homeostasis and glycolysis in ALS pathogenesis. Additionally, we compared our dataset with publicly available data for the SALS spinal cord, and show a high correlation of changes linked to the diseased state in the SALS motor cortex. In an analogous comparison with data for the Alzheimer's disease hippocampus we demonstrate a low correlation of global changes and a moderate correlation for changes specifically linked to the SALS diseased state. CONCLUSION Gene and sample numbers investigated allow pathway- and gene-based analyses by established error-correction methods, drawing a molecular portrait of the ALS motor cortex that faithfully represents many known disease features and uncovers several novel aspects of ALS pathology. Contrary to expectations for a tissue under oxidative stress, nuclear-encoded mitochondrial genes are uniformly down-regulated. Moreover, the down-regulation of mitochondrial and glycolytic genes implies a combined reduction of mitochondrial and cytoplasmic energy supply, with a possible role in the death of ALS motoneurons. Identifying candidate genes exclusively expressed in non-neuronal cells, we also highlight the importance of these cells in disease development in the motor cortex. Notably, some pathways and candidate genes identified by this study are direct or indirect targets of medication already applied to unrelated illnesses and point the way towards the rapid development of effective symptomatic ALS therapies.
Collapse
|
47
|
Caeyenberghs K, Balschun D, Roces DP, Schwake M, Saftig P, D'Hooge R. Multivariate neurocognitive and emotional profile of a mannosidosis murine model for therapy assessment. Neurobiol Dis 2006; 23:422-32. [PMID: 16766199 DOI: 10.1016/j.nbd.2006.03.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2005] [Revised: 03/07/2006] [Accepted: 03/23/2006] [Indexed: 11/24/2022] Open
Abstract
alpha-Mannosidosis is a lysosomal storage disorder caused by lysosomal alpha-mannosidase (LAMAN) deficiency that leads to neurocognitive dysfunctions, psychotic symptoms and emotional changes in human patients. A murine mannosidosis model, LAMAN-deficient mice, was examined on a behavioral task battery that included test for neuromotor, exploratory and neurocognitive (spatial learning and memory) abilities, and multivariate statistical analyses were used to identify behavioral and neurocognitive domains that are most heavily affected by LAMAN deficiency. In addition, we further investigated synaptic plasticity recordings on hippocampal slices that may relate to these behavioral alterations. Correlation analysis revealed significant intra- and intertask correlations and factor analysis that included all 21 behavioral variables identified three main factors (exploration/emotionality, locomotion and learning/memory abilities). Significant correlations were observed between genotype, and factor 1 (exploration/emotionality) and factor 3 (learning/memory abilities). Discriminant function analysis showed that "path length in the open field test" and "time spent in the target quadrant during the water maze probe trial" were the most decisive variables to distinguish between the genotypes. We therefore suggest that these variables would be especially important in forthcoming therapy assessment experiments using this murine mannosidosis model. LAMAN-deficient mice displayed severe changes in synaptic plasticity, which may have contributed to the neurocognitive impairments observed. The present report further shows that targeted deletion of the LAMAN gene in mice mimics many aspects of human alpha-mannosidosis, and these data provide a basis for future therapeutic experiments.
Collapse
Affiliation(s)
- Karen Caeyenberghs
- Laboratory of Biological Psychology, Department of Psychology, KU Leuven, Belgium
| | | | | | | | | | | |
Collapse
|
48
|
Occurrence and co-localization of amyloid beta-protein and apolipoprotein E in perivascular drainage channels of wild-type and APP-transgenic mice. Neurobiol Aging 2006; 28:1221-30. [PMID: 16815595 DOI: 10.1016/j.neurobiolaging.2006.05.029] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2005] [Revised: 05/18/2006] [Accepted: 05/25/2006] [Indexed: 11/26/2022]
Abstract
The deposition of the amyloid beta-protein (Abeta) is a hallmark of Alzheimer's disease (AD). One reason for Abeta-accumulation and deposition in the brain may be an altered drainage along perivascular channels. Extracellular fluid is drained from the brain towards the cervical lymph nodes via perivascular channels. The perivascular space around cerebral arteries is the morphological correlative of these drainage channels. Here, we show that Abeta is immunohistochemically detectable within the perivascular space of 25 months old wild-type and amyloid precursor protein (APP)-transgenic mice harboring the Swedish double mutation driven by a neuron specific promoter. Only small amounts of Abeta can be detected immunohistochemically in the perivascular space of wild-type mice. Cerebrovascular and parenchymal Abeta-deposits were absent. In APP-transgenic mice, large amounts of Abeta were found in the perivascular drainage channels accompanied with cerebrovascular and parenchymal Abeta-deposition. The apolipoprotein E (apoE) immunostaining within the perivascular channels did not vary between wild-type and APP-transgenic mice. Almost 100% of the area that represents the perivascular space was stained with an antibody directed against apoE. Here, Abeta co-localized with apoE indicating an involvement of apoE in the perivascular clearance of Abeta. Fibrillar congophilic amyloid was not seen in wild-type mice. In APP-transgenic animals, congophilic fibrillar amyloid material was seen in the wall of cerebral blood vessels but not in the perivascular space. In conclusion, our results suggest that non-fibrillar forms of Abeta are drained along perivascular channels and that apoE is presumably involved in this clearance mechanism. Overloading such a clearance mechanism in APP-transgenic mice appears to result in insufficient Abeta-clearance, increased Abeta-levels in the brain and the perivascular drainage channels, and finally in Abeta-deposition. In so doing, our results strengthen the hypothesis that an alteration of perivascular drainage supports Abeta-deposition and the development of AD.
Collapse
|
49
|
Lord A, Kalimo H, Eckman C, Zhang XQ, Lannfelt L, Nilsson LNG. The Arctic Alzheimer mutation facilitates early intraneuronal Aβ aggregation and senile plaque formation in transgenic mice. Neurobiol Aging 2006; 27:67-77. [PMID: 16298242 DOI: 10.1016/j.neurobiolaging.2004.12.007] [Citation(s) in RCA: 180] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2004] [Accepted: 12/20/2004] [Indexed: 11/27/2022]
Abstract
The Arctic mutation (APP E693G) is unique, since it is located within the amyloid-beta (Abeta) sequence and leads to Alzheimer's disease (AD). Arctic Abeta peptides more easily form Abeta protofibrils in vitro, but little is known about the pathogenic mechanism of the Arctic mutation in vivo. Here, we analyzed APP transgenic mice with both the Swedish and Arctic mutations (tg-APPArcSwe) and transgenic mice with the Swedish mutation alone (tg-APPSwe). Intense intraneuronal Abeta-immunoreactive staining was present in young tg-APPArcSwe mice, but not in tg-APPSwe mice. Intracellular Abeta aggregates in tg-APPArcSwe were strongly stained by antibodies recognizing the N-terminus of Abeta, while those recognizing the C-terminus of Abeta stained weakly. The Abeta aggregates inside neurons increased with age and predated extracellular Abeta deposition in both tg-APPArcSwe and tg-APPSwe mice. Senile plaque deposition was markedly accelerated in tg-APPArcSwe mice, as compared to tg-APPSwe mice. We conclude that the Arctic mutation causes AD by facilitating amyloidosis through early accumulation of intracellular Abeta aggregates in association with a rapid onset of senile plaque deposition.
Collapse
Affiliation(s)
- Anna Lord
- Department of Public Health and Caring Sciences, Geriatrics, Uppsala University, Dag Hammarskjölds Väg 20, SE-751 85 Uppsala, Sweden
| | | | | | | | | | | |
Collapse
|
50
|
Baker C, Nielsen HM, Minthon L, Wright HT, Chappell S, Okyere J, May S, Morgan K, Kalsheker N, Janciauskiene SM. Effects of Alzheimer's peptide and alpha1-antichymotrypsin on astrocyte gene expression. Neurobiol Aging 2005; 28:51-61. [PMID: 16364502 DOI: 10.1016/j.neurobiolaging.2005.10.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2005] [Revised: 10/20/2005] [Accepted: 10/21/2005] [Indexed: 01/08/2023]
Abstract
We employed gene array technology to investigate the effects of alpha1-antichymotrypsin (ACT), soluble or fibrillar Alzheimer's peptide (Abeta(1-42)) alone and the combination of ACT/Abeta(1-42) on human astrocytes. Using a 1.2-fold change as significance threshold, 398 astrocyte genes showed altered expression in response to these treatments compared to controls. Of the 276 genes affected by the ACT/soluble Abeta(1-42) combination, 195 (70.6%) were suppressed. The ACT/fibrillar Abeta(1-42) combination affected expression of 64 genes of which 58 (90.5%) were up-regulated. The most prominent gene expression changes in response to the ACT/soluble Abeta(1-42), were the down-regulation of at least 60 genes involved in transcription, signal transduction, apoptosis and neurogenesis. The ACT/fibril Abeta(1-42) increased the expression of genes involved in transcription regulation and signal transduction. Surprisingly, gene expression of astrocytes exposed to soluble or fibrillar Abeta(1-42) alone was largely unaffected. Thus, the molecular forms generated by the combination of ACT/Abeta(1-42) alter expression of astrocyte genes more profoundly in breadth and magnitude than soluble or fibrillar Abeta(1-42) alone, suggesting that pathogenic effects of Abeta(1-42) may occur as a consequence of its association with other proteins.
Collapse
Affiliation(s)
- Crystal Baker
- Institute of Genetics, School of Molecular Medical Sciences, University of Nottingham, Nottingham NG7 2UH, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|