1
|
Zhao H, Ji Q, Wu Z, Wang S, Ren J, Yan K, Wang Z, Hu J, Chu Q, Hu H, Cai Y, Wang Q, Huang D, Ji Z, Li J, Belmonte JCI, Song M, Zhang W, Qu J, Liu GH. Destabilizing heterochromatin by APOE mediates senescence. NATURE AGING 2022; 2:303-316. [PMID: 35368774 DOI: 10.1038/s43587-022-00186-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 02/01/2022] [Indexed: 04/30/2023]
Abstract
Apolipoprotein E (APOE) is a component of lipoprotein particles that function in the homeostasis of cholesterol and other lipids. Although APOE is genetically associated with human longevity and Alzheimer's disease, its mechanistic role in aging is largely unknown. Here, we used human genetic, stress-induced and physiological cellular aging models to explore APOE-driven processes in stem cell homeostasis and aging. We report that in aged human mesenchymal progenitor cells (MPCs), APOE accumulation is a driver for cellular senescence. By contrast, CRISPR-Cas9-mediated deletion of APOE endows human MPCs with resistance to cellular senescence. Mechanistically, we discovered that APOE functions as a destabilizer for heterochromatin. Specifically, increased APOE leads to the degradation of nuclear lamina proteins and a heterochromatin-associated protein KRAB-associated protein 1 via the autophagy-lysosomal pathway, thereby disrupting heterochromatin and causing senescence. Altogether, our findings uncover a role of APOE as an epigenetic mediator of senescence and provide potential targets to ameliorate aging-related diseases.
Collapse
Affiliation(s)
- Hongkai Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Science and Technology of China, Hefei, China
| | - Qianzhao Ji
- University of the Chinese Academy of Sciences, Beijing, China
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Zeming Wu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Si Wang
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
- Chongqing Renji Hospital, University of the Chinese Academy of Sciences, Chongqing, China
| | - Jie Ren
- University of the Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Chinese Academy of Sciences Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- China National Center for Bioinformation, Beijing, China
| | - Kaowen Yan
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Zehua Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Jianli Hu
- University of the Chinese Academy of Sciences, Beijing, China
- Chinese Academy of Sciences Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- China National Center for Bioinformation, Beijing, China
| | - Qun Chu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Huifang Hu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Yusheng Cai
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Qiaoran Wang
- University of the Chinese Academy of Sciences, Beijing, China
- Chinese Academy of Sciences Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- China National Center for Bioinformation, Beijing, China
| | - Daoyuan Huang
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Zhejun Ji
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Jingyi Li
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | | | - Moshi Song
- University of the Chinese Academy of Sciences, Beijing, China.
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
| | - Weiqi Zhang
- University of the Chinese Academy of Sciences, Beijing, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
- Chinese Academy of Sciences Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China.
- China National Center for Bioinformation, Beijing, China.
| | - Jing Qu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- University of Science and Technology of China, Hefei, China.
- University of the Chinese Academy of Sciences, Beijing, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
| | - Guang-Hui Liu
- University of the Chinese Academy of Sciences, Beijing, China.
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China.
- Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
2
|
Troutwine BR, Hamid L, Lysaker CR, Strope TA, Wilkins HM. Apolipoprotein E and Alzheimer's disease. Acta Pharm Sin B 2022; 12:496-510. [PMID: 35256931 PMCID: PMC8897057 DOI: 10.1016/j.apsb.2021.10.002] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/30/2021] [Accepted: 10/07/2021] [Indexed: 12/14/2022] Open
Abstract
Genetic variation in apolipoprotein E (APOE) influences Alzheimer's disease (AD) risk. APOE ε4 alleles are the strongest genetic risk factor for late onset sporadic AD. The AD risk is dose dependent, as those carrying one APOE ε4 allele have a 2-3-fold increased risk, while those carrying two ε4 alleles have a 10-15-fold increased risk. Individuals carrying APOE ε2 alleles have lower AD risk and those carrying APOE ε3 alleles have neutral risk. APOE is a lipoprotein which functions in lipid transport, metabolism, and inflammatory modulation. Isoform specific effects of APOE within the brain include alterations to Aβ, tau, neuroinflammation, and metabolism. Here we review the association of APOE with AD, the APOE isoform specific effects within brain and periphery, and potential therapeutics.
Collapse
Affiliation(s)
- Benjamin R. Troutwine
- Department of Neurology University of Kansas Medical Center, Kansas City, KS 66160, USA
- University of Kansas Alzheimer's Disease Center, Kansas City, KS 66160, USA
| | - Laylan Hamid
- University of Kansas Alzheimer's Disease Center, Kansas City, KS 66160, USA
| | - Colton R. Lysaker
- University of Kansas Alzheimer's Disease Center, Kansas City, KS 66160, USA
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Taylor A. Strope
- University of Kansas Alzheimer's Disease Center, Kansas City, KS 66160, USA
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Heather M. Wilkins
- Department of Neurology University of Kansas Medical Center, Kansas City, KS 66160, USA
- University of Kansas Alzheimer's Disease Center, Kansas City, KS 66160, USA
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
3
|
Copper, Iron, Selenium and Lipo-Glycemic Dysmetabolism in Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms22179461. [PMID: 34502369 PMCID: PMC8431716 DOI: 10.3390/ijms22179461] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 12/18/2022] Open
Abstract
The aim of the present review is to discuss traditional hypotheses on the etiopathogenesis of Alzheimer's disease (AD), as well as the role of metabolic-syndrome-related mechanisms in AD development with a special focus on advanced glycation end-products (AGEs) and their role in metal-induced neurodegeneration in AD. Persistent hyperglycemia along with oxidative stress results in increased protein glycation and formation of AGEs. The latter were shown to possess a wide spectrum of neurotoxic effects including increased Aβ generation and aggregation. In addition, AGE binding to receptor for AGE (RAGE) induces a variety of pathways contributing to neuroinflammation. The existing data also demonstrate that AGE toxicity seems to mediate the involvement of copper (Cu) and potentially other metals in AD pathogenesis. Specifically, Cu promotes AGE formation, AGE-Aβ cross-linking and up-regulation of RAGE expression. Moreover, Aβ glycation was shown to increase prooxidant effects of Cu through Fenton chemistry. Given the role of AGE and RAGE, as well as metal toxicity in AD pathogenesis, it is proposed that metal chelation and/or incretins may slow down oxidative damage. In addition, selenium (Se) compounds seem to attenuate the intracellular toxicity of the deranged tau and Aβ, as well as inhibiting AGE accumulation and metal-induced neurotoxicity.
Collapse
|
4
|
Wang H, Chen MH, Chen W, Zhang JG, Qin SC. Roles and mechanisms of phospholipid transfer protein in the development of Alzheimer's disease. Psychogeriatrics 2021; 21:659-667. [PMID: 33851473 DOI: 10.1111/psyg.12685] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 02/25/2021] [Accepted: 03/02/2021] [Indexed: 01/20/2023]
Abstract
Phospholipid transfer protein (PLTP) is a complex glycosylated protein that mediates the transfer of phospholipids, unesterified cholesterol, diacylglycerides, specific apolipoproteins, and tocopherols between different classes of lipoproteins as well as between lipoproteins and cells. Many studies have associated PLTP with a variety of lipid metabolic diseases. However, recent studies have indicated that PLTP is highly expressed in the brain of vertebrate and may be related to many central nervous system diseases, such as Alzheimer's disease. Here, we review the data and report the role and mechanisms PLTP in Alzheimer's disease.
Collapse
Affiliation(s)
- Hao Wang
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai'an, China
| | - Mei-Hua Chen
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai'an, China
| | - Wei Chen
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai'an, China
| | - Ji-Guo Zhang
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai'an, China
| | - Shu-Cun Qin
- Key Laboratory of Atherosclerosis in Universities of Shandong; Institute of Atherosclerosis, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai'an, China
| |
Collapse
|
5
|
Ringland C, Schweig JE, Eisenbaum M, Paris D, Ait-Ghezala G, Mullan M, Crawford F, Abdullah L, Bachmeier C. MMP9 modulation improves specific neurobehavioral deficits in a mouse model of Alzheimer's disease. BMC Neurosci 2021; 22:39. [PMID: 34034683 PMCID: PMC8152085 DOI: 10.1186/s12868-021-00643-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 05/10/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Matrix metallopeptidase 9 (MMP9) has been implicated in a variety of neurological disorders, including Alzheimer's disease (AD), where MMP9 levels are elevated in the brain and cerebrovasculature. Previously our group demonstrated apolipoprotein E4 (apoE4) was less efficient in regulating MMP9 activity in the brain than other apoE isoforms, and that MMP9 inhibition facilitated beta-amyloid (Aβ) elimination across the blood-brain barrier (BBB) METHODS: In the current studies, we evaluated the impact of MMP9 modulation on Aβ disposition and neurobehavior in AD using two approaches, (1) pharmacological inhibition of MMP9 with SB-3CT in apoE4 x AD (E4FAD) mice, and (2) gene deletion of MMP9 in AD mice (MMP9KO/5xFAD) RESULTS: Treatment with the MMP9 inhibitor SB-3CT in E4FAD mice led to reduced anxiety compared to placebo using the elevated plus maze. Deletion of the MMP9 gene in 5xFAD mice also reduced anxiety using the open field test, in addition to improving sociability and social recognition memory, particularly in male mice, as assessed through the three-chamber task, indicating certain behavioral alterations in AD may be mediated by MMP9. However, neither pharmacological inhibition of MMP9 or gene deletion of MMP9 affected spatial learning or memory in the AD animals, as determined through the radial arm water maze. Moreover, the effect of MMP9 modulation on AD neurobehavior was not due to changes in Aβ disposition, as both brain and plasma Aβ levels were unchanged in the SB-3CT-treated E4FAD animals and MMP9KO/AD mice compared to their respective controls. CONCLUSIONS In total, while MMP9 inhibition did improve specific neurobehavioral deficits associated with AD, such as anxiety and social recognition memory, modulation of MMP9 did not alter spatial learning and memory or Aβ tissue levels in AD animals. While targeting MMP9 may represent a therapeutic strategy to mitigate aspects of neurobehavioral decline in AD, further work is necessary to understand the nature of the relationship between MMP9 activity and neurological dysfunction.
Collapse
Affiliation(s)
- Charis Ringland
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA
- The Open University, Milton Keynes, UK
| | | | - Maxwell Eisenbaum
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA
- The Open University, Milton Keynes, UK
| | - Daniel Paris
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA
| | - Ghania Ait-Ghezala
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA
- The Open University, Milton Keynes, UK
| | - Michael Mullan
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA
- The Open University, Milton Keynes, UK
| | - Fiona Crawford
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA
- The Open University, Milton Keynes, UK
- James A. Haley Veterans' Hospital, Tampa, FL, USA
| | - Laila Abdullah
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA
- The Open University, Milton Keynes, UK
- James A. Haley Veterans' Hospital, Tampa, FL, USA
| | - Corbin Bachmeier
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA.
- The Open University, Milton Keynes, UK.
- Bay Pines VA Healthcare System, Bay Pines, FL, USA.
| |
Collapse
|
6
|
Soares Martins T, Marçalo R, Ferreira M, Vaz M, Silva RM, Martins Rosa I, Vogelgsang J, Wiltfang J, da Cruz e Silva OAB, Henriques AG. Exosomal Aβ-Binding Proteins Identified by "In Silico" Analysis Represent Putative Blood-Derived Biomarker Candidates for Alzheimer´s Disease. Int J Mol Sci 2021; 22:ijms22083933. [PMID: 33920336 PMCID: PMC8070602 DOI: 10.3390/ijms22083933] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 04/07/2021] [Indexed: 12/17/2022] Open
Abstract
The potential of exosomes as biomarker resources for diagnostics and even for therapeutics has intensified research in the field, including in the context of Alzheimer´s disease (AD). The search for disease biomarkers in peripheral biofluids is advancing mainly due to the easy access it offers. In the study presented here, emphasis was given to the bioinformatic identification of putative exosomal candidates for AD. The exosomal proteomes of cerebrospinal fluid (CSF), serum and plasma, were obtained from three databases (ExoCarta, EVpedia and Vesiclepedia), and complemented with additional exosomal proteins already associated with AD but not found in the databases. The final biofluids’ proteomes were submitted to gene ontology (GO) enrichment analysis and the exosomal Aβ-binding proteins that can constitute putative candidates were identified. Among these candidates, gelsolin, a protein known to be involved in inhibiting Abeta fibril formation, was identified, and it was tested in human samples. The levels of this Aβ-binding protein, with anti-amyloidogenic properties, were assessed in serum-derived exosomes isolated from controls and individuals with dementia, including AD cases, and revealed altered expression patterns. Identification of potential peripheral biomarker candidates for AD may be useful, not only for early disease diagnosis but also in drug trials and to monitor disease progression, allowing for a timely therapeutic intervention, which will positively impact the patient’s quality of life.
Collapse
Affiliation(s)
- Tânia Soares Martins
- Neurosciences and Signalling Group, Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro (UA), 3810-193 Aveiro, Portugal; (T.S.M.); (R.M.); (M.F.); (M.V.); (I.M.R.); (J.W.); (O.C.S.)
| | - Rui Marçalo
- Neurosciences and Signalling Group, Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro (UA), 3810-193 Aveiro, Portugal; (T.S.M.); (R.M.); (M.F.); (M.V.); (I.M.R.); (J.W.); (O.C.S.)
| | - Maria Ferreira
- Neurosciences and Signalling Group, Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro (UA), 3810-193 Aveiro, Portugal; (T.S.M.); (R.M.); (M.F.); (M.V.); (I.M.R.); (J.W.); (O.C.S.)
| | - Margarida Vaz
- Neurosciences and Signalling Group, Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro (UA), 3810-193 Aveiro, Portugal; (T.S.M.); (R.M.); (M.F.); (M.V.); (I.M.R.); (J.W.); (O.C.S.)
| | - Raquel M. Silva
- Center for Interdisciplinary Research in Health (CIIS), Faculdade de Medicina Dentária, Universidade Católica Portuguesa, Estrada da Circunvalação, 3504-505 Viseu, Portugal;
| | - Ilka Martins Rosa
- Neurosciences and Signalling Group, Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro (UA), 3810-193 Aveiro, Portugal; (T.S.M.); (R.M.); (M.F.); (M.V.); (I.M.R.); (J.W.); (O.C.S.)
| | - Jonathan Vogelgsang
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen (UMG), Georg-August University, Von-Siebold-Str. 5, 37075 Goettingen, Germany;
- Translational Neuroscience Laboratory, McLean Hospital, Harvard Medical School, Belmont, MA 02478, USA
| | - Jens Wiltfang
- Neurosciences and Signalling Group, Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro (UA), 3810-193 Aveiro, Portugal; (T.S.M.); (R.M.); (M.F.); (M.V.); (I.M.R.); (J.W.); (O.C.S.)
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen (UMG), Georg-August University, Von-Siebold-Str. 5, 37075 Goettingen, Germany;
- German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Str. 3a, 37075 Goettingen, Germany
| | - Odete A. B. da Cruz e Silva
- Neurosciences and Signalling Group, Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro (UA), 3810-193 Aveiro, Portugal; (T.S.M.); (R.M.); (M.F.); (M.V.); (I.M.R.); (J.W.); (O.C.S.)
| | - Ana Gabriela Henriques
- Neurosciences and Signalling Group, Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro (UA), 3810-193 Aveiro, Portugal; (T.S.M.); (R.M.); (M.F.); (M.V.); (I.M.R.); (J.W.); (O.C.S.)
- Correspondence:
| |
Collapse
|
7
|
Tsai CL, Erickson KI, Sun HS, Kuo YM, Pai MC. A cross-sectional examination of a family history of Alzheimer's disease and ApoE epsilon 4 on physical fitness, molecular biomarkers, and neurocognitive performance. Physiol Behav 2020; 230:113268. [PMID: 33383402 DOI: 10.1016/j.physbeh.2020.113268] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 10/30/2020] [Accepted: 11/21/2020] [Indexed: 10/22/2022]
Abstract
PURPOSE The present study examined whether the ɛ4 allele of the apolipoprotein E (ApoE) gene impacts molecular biomarkers and neurocognitive performance among individuals at genetic risk for developing Alzheimer's disease (AD). The correlations between physical fitness and molecular/neurocognitive indices were also explored. METHODS Fasting blood samples were collected from 162 individuals with a family history of AD (ADFH). There were twenty-two carriers of the ApoE-4 variant (ApoE-4 group). For comparison purposes we randomly selected 22 non-ɛ4 carriers (non-ApoE-4 group) from the ADFH individuals. Circulating inflammatory cytokines (e.g., TNF-α, IL-1β, IL-6, IL-8, and IL-15), neuroprotective growth factors (e.g., BDNF, IGF-1, IGF-2, VEGF, and FGF-2), and Amyloid-β peptides (e.g., Aβ1-40 and Aβ1-42), neurocognitive performance [e.g., behavior and brain even-related potentials (ERP)] during a task-switching paradigm, as well as physical fitness scores were measured. RESULTS The ApoE-4 group relative to the non-ApoE-4 group was similar with respect to molecular biomarkers, physical fitness, and most measures of neurocognitive performance. However, ADFH individuals that were ɛ4 carriers exhibited significantly higher local switching accuracy costs, worse accuracy as well as smaller ERP P3 amplitudes for the memory-switching condition. Importantly, cardiorespiratory fitness levels were significantly correlated with accuracy for most task-switching conditions, and levels of BDNF, Aβ1-40, and Aβ1-42 collapsed across the two groups even when controlling for the age co-variable, while the ApoE-4 group revealed similar pattern of results. CONCLUSIONS These data suggest that individuals with ADFH that were carriers of the ApoE-4 variant performed worse on the task-switching paradigm and that this could be due to compromised task-set and memory updating processes. Physical exercise interventions aimed to enhance cardiorespiratory fitness levels could be a potential AD prevention strategy for ameliorating cognitive function and reducing the accumulation of the Aβ peptides in this high risk group.
Collapse
Affiliation(s)
- Chia-Liang Tsai
- Institute of Physical Education, Health and Leisure Studies, National Cheng Kung University, Taiwan.
| | - Kirk I Erickson
- Department of Psychology, University of Pittsburgh, USA; Discipline of Exercise Science, College of Science, Health, Engineering and Education, Murdoch University, Western Australia
| | - H-Sunny Sun
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Taiwan
| | - Yu-Min Kuo
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Taiwan
| | - Ming-Chyi Pai
- Division of Behavioral Neurology, Department of Neurology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Taiwan; Alzheimer's Disease Research Center, National Cheng Kung University Hospital, Taiwan.
| |
Collapse
|
8
|
Ojo J, Eisenbaum M, Shackleton B, Lynch C, Joshi U, Saltiel N, Pearson A, Ringland C, Paris D, Mouzon B, Mullan M, Crawford F, Bachmeier C. Mural cell dysfunction leads to altered cerebrovascular tau uptake following repetitive head trauma. Neurobiol Dis 2020; 150:105237. [PMID: 33383188 PMCID: PMC8170787 DOI: 10.1016/j.nbd.2020.105237] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 12/07/2020] [Accepted: 12/26/2020] [Indexed: 12/29/2022] Open
Abstract
A pathological characteristic of repetitive traumatic brain injury (TBI) is the deposition of hyperphosphorylated and aggregated tau species in the brain and increased levels of extracellular monomeric tau are believed to play a role in the pathogenesis of neurodegenerative tauopathies. The pathways by which extracellular tau is eliminated from the brain, however, remains elusive. The purpose of this study was to examine tau uptake by cerebrovascular cells and the effect of TBI on these processes. We found monomeric tau interacts with brain vascular mural cells (pericytes and smooth muscle cells) to a greater extent than other cerebrovascular cells, indicating mural cells may contribute to the elimination of extracellular tau, as previously described for other solutes such as beta-amyloid. Consistent with other neurodegenerative disorders, we observed a progressive decline in cerebrovascular mural cell markers up to 12 months post-injury in a mouse model of repetitive mild TBI (r-mTBI) and human TBI brain specimens, when compared to control. These changes appear to reflect mural cell degeneration and not cellular loss as no difference in the mural cell population was observed between r-mTBI and r-sham animals as determined through flow cytometry. Moreover, freshly isolated r-mTBI cerebrovessels showed reduced tau uptake at 6 and 12 months post-injury compared to r-sham animals, which may be the result of diminished cerebrovascular endocytosis, as caveolin-1 levels were significantly decreased in mouse r-mTBI and human TBI cerebrovessels compared to their respective controls. Further emphasizing the interaction between mural cells and tau, similar reductions in mural cell markers, tau uptake, and caveolin-1 were observed in cerebrovessels from transgenic mural cell-depleted animals. In conclusion, our studies indicate repeated injuries to the brain causes chronic mural cell degeneration, reducing the caveolar-mediated uptake of tau by these cells. Alterations in tau uptake by vascular mural cells may contribute to tau deposition in the brain following head trauma and could represent a novel therapeutic target for TBI or other neurodegenerative disorders.
Collapse
Affiliation(s)
- Joseph Ojo
- The Roskamp Institute, Sarasota, FL, USA; The Open University, Milton Keynes, UK
| | - Max Eisenbaum
- The Roskamp Institute, Sarasota, FL, USA; The Open University, Milton Keynes, UK
| | - Ben Shackleton
- The Roskamp Institute, Sarasota, FL, USA; The Open University, Milton Keynes, UK
| | - Cillian Lynch
- The Roskamp Institute, Sarasota, FL, USA; The Open University, Milton Keynes, UK
| | - Utsav Joshi
- The Roskamp Institute, Sarasota, FL, USA; The Open University, Milton Keynes, UK
| | | | - Andrew Pearson
- The Roskamp Institute, Sarasota, FL, USA; The Open University, Milton Keynes, UK
| | - Charis Ringland
- The Roskamp Institute, Sarasota, FL, USA; The Open University, Milton Keynes, UK
| | - Daniel Paris
- The Roskamp Institute, Sarasota, FL, USA; The Open University, Milton Keynes, UK
| | - Benoit Mouzon
- The Roskamp Institute, Sarasota, FL, USA; The Open University, Milton Keynes, UK
| | - Michael Mullan
- The Roskamp Institute, Sarasota, FL, USA; The Open University, Milton Keynes, UK
| | - Fiona Crawford
- The Roskamp Institute, Sarasota, FL, USA; The Open University, Milton Keynes, UK; James A. Haley Veterans' Hospital, Tampa, FL, USA
| | - Corbin Bachmeier
- The Roskamp Institute, Sarasota, FL, USA; The Open University, Milton Keynes, UK; Bay Pines VA Healthcare System, Bay Pines, FL, USA.
| |
Collapse
|
9
|
Wünsch A, Mulac D, Langer K. Lipoprotein imitating nanoparticles: Lecithin coating binds ApoE and mediates non-lysosomal uptake leading to transcytosis over the blood-brain barrier. Int J Pharm 2020; 589:119821. [DOI: 10.1016/j.ijpharm.2020.119821] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/14/2020] [Accepted: 08/24/2020] [Indexed: 12/29/2022]
|
10
|
Zhang QZ, Xue AY, Wei W, Pang AM, Cao LN, Liu F. Deletion of P110δ promotes the development of myocarditis in ApoE‑deficient mice by increasing mononuclear cell peritoneal infiltration. Mol Med Rep 2020; 22:3629-3634. [PMID: 33000201 PMCID: PMC7533459 DOI: 10.3892/mmr.2020.11451] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 04/06/2018] [Indexed: 11/06/2022] Open
Abstract
Phosphoinositide 3-kinase catalytic subunit δ isoform (P110δ) is mainly expressed in white blood cells. It is involved in T and B lymphocyte differentiation, maturation and the neutrophil chemotaxis process. Apolipoprotein E (ApoE) is an arginine‑rich alkaline protein, which is present in plasma chylomicron, low‑density lipoprotein and very low‑density lipoprotein. The present study aimed to determine the effects of P110δ deletion on myocarditis in ApoE‑/‑ mice. A mouse model of ApoE and P110δ double deletion was initially constructed; hematoxylin and eosin (H&E) staining was performed to detect the histological alterations in the mouse myocardium. Systolic and diastolic alterations, and alterations in the left ventricular fractional shortening (LVFS) and left ventricular ejection fraction (LVEF) were examined by electrocardiogram. Blood cell of ApoE and P110δ double mice was used to detect changes in white blood cells and monocytes. Western blotting was used to detect the expression levels of apoptosis‑associated proteins, whereas flow cytometry was used to detect the percentage of apoptosis. Morphological alterations in myocardial cells were observed under a microscope. The results of polymerase chain reaction demonstrated that double deletion mice were successfully constructed. H&E staining revealed that cells in the ApoE‑/‑ mice were spindle‑shaped; however, the nuclei were smaller in the double deletion mice. There was no change in cardiac contraction in normal mice; however, in double deletion mice, the systolic and diastolic contractions were markedly reduced. LVFS and LVEF were decreased compared with in the control group. Blood cell analysis indicated that the content of white blood cells and monocytes in the experimental group was significantly higher than that in the control group. Western blotting demonstrated that the expression levels of apoptotic proteins in double deletion mice were significantly higher compared with in the control group. Flow cytometry revealed that the apoptotic ratio was increased in double deletion mice compared with in the control group (42 vs. 21%). These findings suggested that deletion of P110δ may induce monocyte peritoneal infiltration and increase apoptosis, thus promoting the development of myocarditis.
Collapse
Affiliation(s)
- Qi-Zhi Zhang
- Out‑Patient Department, The Second Hospital of Shandong University, Jinan, Shandong 250000, P.R. China
| | - Ai-Ying Xue
- Department of Cardiology, The Second Hospital of Shandong University, Jinan, Shandong 250000, P.R. China
| | - Wei Wei
- Nuclear Medicine, The Second Hospital of Shandong University, Jinan, Shandong 250000, P.R. China
| | - Ai-Min Pang
- Out‑Patient Department, The Second Hospital of Shandong University, Jinan, Shandong 250000, P.R. China
| | - Li-Na Cao
- Department of Cardiology, The Second Hospital of Shandong University, Jinan, Shandong 250000, P.R. China
| | - Fang Liu
- Department of Cardiology, The Second Hospital of Shandong University, Jinan, Shandong 250000, P.R. China
| |
Collapse
|
11
|
Ringland C, Schweig JE, Paris D, Shackleton B, Lynch CE, Eisenbaum M, Mullan M, Crawford F, Abdullah L, Bachmeier C. Apolipoprotein E isoforms differentially regulate matrix metallopeptidase 9 function in Alzheimer's disease. Neurobiol Aging 2020; 95:56-68. [PMID: 32758917 DOI: 10.1016/j.neurobiolaging.2020.06.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 06/05/2020] [Accepted: 06/25/2020] [Indexed: 01/10/2023]
Abstract
Apolipoprotein E (APOE) has been shown to influence amyloid-β (Aβ) clearance from the brain in an isoform-specific manner. Our prior work showed that Aβ transit across the blood-brain-barrier was reduced by apoE4, compared to other apoE isoforms, due to elevated lipoprotein receptor shedding in brain endothelia. Recently, we demonstrated that matrix metallopeptidase 9 (MMP-9) induces lipoprotein receptor proteolysis in an apoE isoform-dependent manner, which impacts Aβ elimination from the brain. The current studies interrogated the relationship between apoE and MMP-9 and found that apoE impacted proMMP-9 cellular secretion from brain endothelia (apoE2 < apoE3 = apoE4). In a cell-free assay, apoE dose-dependently reduced MMP-9 activity, with apoE4 showing a significantly weaker ability to inhibit MMP-9 function than apoE2 or apoE3. Finally, we observed elevated MMP-9 expression and activity in the cerebrovasculature of both human and animal AD brain specimens with an APOE4 genotype. Collectively, these findings suggest a role for apoE in regulating MMP-9 disposition and may describe the effect of apoE4 on Aβ pathology in the AD brain.
Collapse
Affiliation(s)
- Charis Ringland
- The Roskamp Institute, Sarasota, FL, USA; The Open University, Milton Keynes, UK.
| | | | | | | | | | - Maxwell Eisenbaum
- The Roskamp Institute, Sarasota, FL, USA; The Open University, Milton Keynes, UK
| | - Michael Mullan
- The Roskamp Institute, Sarasota, FL, USA; The Open University, Milton Keynes, UK
| | - Fiona Crawford
- The Roskamp Institute, Sarasota, FL, USA; The Open University, Milton Keynes, UK; James A. Haley Veterans' Hospital, Tampa, FL, USA
| | - Laila Abdullah
- The Roskamp Institute, Sarasota, FL, USA; The Open University, Milton Keynes, UK; James A. Haley Veterans' Hospital, Tampa, FL, USA
| | - Corbin Bachmeier
- The Roskamp Institute, Sarasota, FL, USA; The Open University, Milton Keynes, UK; Bay Pines VA Healthcare System, Bay Pines, FL, USA
| |
Collapse
|
12
|
Liyanage SI, Weaver DF. Misfolded proteins as a therapeutic target in Alzheimer's disease. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2020; 118:371-411. [PMID: 31928732 DOI: 10.1016/bs.apcsb.2019.08.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
For decades, Alzheimer's Disease (AD) was defined as a disorder of protein misfolding and aggregation. In particular, the extracellular peptide fragment: amyloid-β (Aβ), and the intracellular microtubule-associated protein: tau, were thought to initiate a neurodegenerative cascade which culminated in AD's progressive loss of memory and executive function. As such, both proteins became the focus of intense scrutiny, and served as the principal pathogenic target for hundreds of clinical trials. However, with varying efficacy, none of these investigations produced a disease-modifying therapy - offering patients with AD little recourse aside from transient, symptomatic medications. The near universal failure of clinical trials is unprecedented for a major research discipline. In part, this has motivated an increasing skepticism of the relevance of protein misfolding to AD's etiology. Several recent observations, principally the presence of significant protein pathologies in non-demented seniors, have lent credence to an apparent cursory role for Aβ and tau. Herein, we review both Aβ and tau, examining the processes from their biosynthesis to their pathogenesis and evaluate their vulnerability to medicinal intervention. We further attempt to reconcile the apparent failure of trials with the potential these targets hold. Ultimately, we seek to answer if protein misfolding is a viable platform in the pursuit of a disease-arresting strategy for AD.
Collapse
Affiliation(s)
- S Imindu Liyanage
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Donald F Weaver
- Krembil Research Institute, University Health Network, Toronto, ON, Canada; Departments of Medicine (Neurology), Chemistry and Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
13
|
Gao L, Jiang Y, Wei S, Shang S, Li P, Chen C, Dang L, Wang J, Huo K, Deng M, Wang J, Zhang R, Qu Q. The Level of Plasma Amyloid-β40 Is Correlated with Peripheral Transport Proteins in Cognitively Normal Adults: A Population-Based Cross-Sectional Study. J Alzheimers Dis 2019; 65:951-961. [PMID: 30103331 DOI: 10.3233/jad-180399] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BACKGROUND Transport proteins, soluble low-density lipoprotein receptor-related protein-1 (sLRP1), and soluble receptor of advanced glycation end products (sRAGE), play an important role in the clearance of plasma amyloid-β (Aβ). However, their relationship is not clear. OBJECTIVE The aim was to explore the relationship between plasma levels of sLRP1, sRAGE, and Aβ in a cross-sectional study. METHODS A total of 1,185 cognitively normal participants (age above 40) from a village in the suburbs of Xi'an, China were enrolled from October 8, 2014 to March 30, 2015. Plasma Aβ40, Aβ42, sLRP1, and sRAGE were tested using a commercial ELISA. Apolipoprotein E (APOE) genotyping was conducted using PCR and sequencing. The relationship between plasma levels of sLRP1, sRAGE, and Aβ was analyzed using Pearson's correlation analysis and multiple linear regression. RESULTS In the total population, Log sLRP1 and Log sRAGE were positively correlated with plasma Aβ40 (r= 0.103, p < 0.001; r= 0.064, p = 0.027, respectively), but neither were associated with plasma Aβ42. After multivariable adjustment in the regression model, Log sLRP1 and Log sRAGE were still positively related with plasma Aβ40 (β= 2.969, p < 0.001; β= 1.936, p = 0.017, respectively) but not Aβ42. Furthermore, the positive correlations between transport proteins and plasma Aβ40 remained significant only in APOEɛ4 non-carriers after Pearson's analysis and multiple regression analysis after stratification by gene status. CONCLUSION The concentrations of plasma sLRP1 and sRAGE had a significant impact on the level of plasma Aβ40 in cognitively normal adults, especially in APOEɛ4 non-carriers. However, the mechanism by which the transport proteins are involved in peripheral Aβ clearance and the relationship between transporters and amyloid burden in the brain needs further validation.
Collapse
Affiliation(s)
- Ling Gao
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yu Jiang
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shan Wei
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Suhang Shang
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Pei Li
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Chen Chen
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Liangjun Dang
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jin Wang
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Kang Huo
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Meiying Deng
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jingyi Wang
- Huxian Hospital of Traditional Chinese Medicine, Xi'an, China
| | - Rong Zhang
- Cerebrovascular Laboratory, Institute for Exercise and Environmental Medicine, UT Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Qiumin Qu
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
14
|
Saint-Pol J, Gosselet F. Oxysterols and the NeuroVascular Unit (NVU): A far true love with bright and dark sides. J Steroid Biochem Mol Biol 2019; 191:105368. [PMID: 31026511 DOI: 10.1016/j.jsbmb.2019.04.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 04/10/2019] [Accepted: 04/22/2019] [Indexed: 12/13/2022]
Abstract
The brain is isolated from the whole body by the blood-brain barrier (BBB) which is located in brain microvessel endothelial cells (ECs). Through physical and metabolic properties induced by brain pericytes, astrocytes and neurons (these cells and the ECs referred to as the neurovascular unit (NVU)), the BBB hardly restricts exchanges of molecules between the brain and the bloodstream. Among them, cholesterol exchanges between these two compartments are very limited and occur through the transport of LDLs across the BBB. Oxysterols (mainly 24S and 27-hydroxycholesterol) daily cross the BBB and regulate molecule/cholesterol exchanges via Liver X nuclear Receptors (LXRs). In addition, these oxysterols have been linked to pathological processes in neurodegenerative diseases such as Alzheimer's disease. Here we propose an overview of the actual knowledge concerning oxysterols and the NVU cells in physiological and in Alzheimer's disease.
Collapse
Affiliation(s)
- Julien Saint-Pol
- University of Artois, Blood-Brain Barrier Laboratory (BBB Lab), EA2465, F-62300 Lens, France.
| | - Fabien Gosselet
- University of Artois, Blood-Brain Barrier Laboratory (BBB Lab), EA2465, F-62300 Lens, France
| |
Collapse
|
15
|
Influence of Matrix Metallopeptidase 9 on Beta-Amyloid Elimination Across the Blood-Brain Barrier. Mol Neurobiol 2019; 56:8296-8305. [PMID: 31209784 DOI: 10.1007/s12035-019-01672-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 06/05/2019] [Indexed: 10/26/2022]
Abstract
Lipoprotein receptor transport across the blood-brain barrier (BBB) mediates beta-amyloid (Aβ) accumulation in the brain and may be a contributing factor in Alzheimer's disease (AD) pathogenesis. Lipoprotein receptors are susceptible to proteolytic shedding at the cell surface, which precludes the endocytic transport of ligands. A ligand that closely interacts with the lipoprotein receptors is apolipoprotein E (apoE), which exists as three isoforms (apoE2, apoE3, apoE4). Our prior work showed an inverse relationship between lipoprotein receptor shedding and Aβ transport across the BBB, which was apoE-isoform dependent. To interrogate this further, the current studies investigated an enzyme implicated in lipoprotein receptor shedding, matrix metalloproteinase 9 (MMP9). Treatment with MMP9 dose-dependently elevated lipoprotein receptor shedding in brain endothelial cells and freshly isolated mouse cerebrovessels. Furthermore, treatment with a MMP9 inhibitor (SB-3CT) mitigated Aβ-induced lipoprotein receptor shedding in brain endothelial cells and the brains of apoE4 animals. In terms of BBB transit, SB-3CT treatment increased the transport of Aβ across an in vitro model of the BBB. In vivo, administration of SB-3CT to apoE4 animals significantly enhanced Aβ clearance from the brain to the periphery following intracranial administration of Aβ. The current studies show that MMP9 impacts lipoprotein receptor shedding and Aβ transit across the BBB, in an apoE isoform-specific manner. In total, MMP9 inhibition can facilitate Aβ clearance across the BBB, which could be an effective approach to lowering Aβ levels in the brain and mitigating the AD phenotype, particularly in subjects carrying the apoE4 allele.
Collapse
|
16
|
Goudey B, Fung BJ, Schieber C, Faux NG. A blood-based signature of cerebrospinal fluid Aβ 1-42 status. Sci Rep 2019; 9:4163. [PMID: 30853713 PMCID: PMC6409361 DOI: 10.1038/s41598-018-37149-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 12/03/2018] [Indexed: 12/22/2022] Open
Abstract
It is increasingly recognized that Alzheimer's disease (AD) exists before dementia is present and that shifts in amyloid beta occur long before clinical symptoms can be detected. Early detection of these molecular changes is a key aspect for the success of interventions aimed at slowing down rates of cognitive decline. Recent evidence indicates that of the two established methods for measuring amyloid, a decrease in cerebrospinal fluid (CSF) amyloid β1-42 (Aβ1-42) may be an earlier indicator of Alzheimer's disease risk than measures of amyloid obtained from Positron Emission Tomography (PET). However, CSF collection is highly invasive and expensive. In contrast, blood collection is routinely performed, minimally invasive and cheap. In this work, we develop a blood-based signature that can provide a cheap and minimally invasive estimation of an individual's CSF amyloid status using a machine learning approach. We show that a Random Forest model derived from plasma analytes can accurately predict subjects as having abnormal (low) CSF Aβ1-42 levels indicative of AD risk (0.84 AUC, 0.78 sensitivity, and 0.73 specificity). Refinement of the modeling indicates that only APOEε4 carrier status and four plasma analytes (CGA, Aβ1-42, Eotaxin 3, APOE) are required to achieve a high level of accuracy. Furthermore, we show across an independent validation cohort that individuals with predicted abnormal CSF Aβ1-42 levels transitioned to an AD diagnosis over 120 months significantly faster than those with predicted normal CSF Aβ1-42 levels and that the resulting model also validates reasonably across PET Aβ1-42 status (0.78 AUC). This is the first study to show that a machine learning approach, using plasma protein levels, age and APOEε4 carrier status, is able to predict CSF Aβ1-42 status, the earliest risk indicator for AD, with high accuracy.
Collapse
Affiliation(s)
- Benjamin Goudey
- IBM Research Australia, Carlton, Victoria, Australia
- Centre for Epidemiology and Biostatistics, The University of Melbourne, Parkville, Victoria, Australia
- Department of Computing and Information System, The University of Melbourne, Parkville, Victoria, Australia
| | - Bowen J Fung
- IBM Research Australia, Carlton, Victoria, Australia
- School of Psychological Sciences, University of Melbourne, Parkville, Victoria, Australia
| | | | - Noel G Faux
- IBM Research Australia, Carlton, Victoria, Australia.
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
17
|
Su H, Jin Y, Noroozifar M, Kerman K. Electrochemical Detection of Isoform‐Specific Interaction between Apolipoprotein E and Amyloid‐β. ChemElectroChem 2018. [DOI: 10.1002/celc.201800985] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Han Su
- Department of Physical and Environmental SciencesUniversity of Toronto Scarborough Toronto Ontario M1 C 1 A4 Canada
| | - Yiyun Jin
- Department of Physical and Environmental SciencesUniversity of Toronto Scarborough Toronto Ontario M1 C 1 A4 Canada
| | - Meissam Noroozifar
- Department of ChemistryUniversity of Sistan and Baluchestan Zahedan Iran
| | - Kagan Kerman
- Department of Physical and Environmental SciencesUniversity of Toronto Scarborough Toronto Ontario M1 C 1 A4 Canada
| |
Collapse
|
18
|
Rowland HA, Hooper NM, Kellett KAB. Modelling Sporadic Alzheimer's Disease Using Induced Pluripotent Stem Cells. Neurochem Res 2018; 43:2179-2198. [PMID: 30387070 PMCID: PMC6267251 DOI: 10.1007/s11064-018-2663-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 09/11/2018] [Accepted: 10/15/2018] [Indexed: 12/24/2022]
Abstract
Developing cellular models of sporadic Alzheimer's disease (sAD) is challenging due to the unknown initiator of disease onset and the slow disease progression that takes many years to develop in vivo. The use of human induced pluripotent stem cells (iPSCs) has revolutionised the opportunities to model AD pathology, investigate disease mechanisms and screen potential drugs. The majority of this work has, however, used cells derived from patients with familial AD (fAD) where specific genetic mutations drive disease onset. While these provide excellent models to investigate the downstream pathways involved in neuronal toxicity and ultimately neuronal death that leads to AD, they provide little insight into the causes and mechanisms driving the development of sAD. In this review we compare the data obtained from fAD and sAD iPSC-derived cell lines, identify the inconsistencies that exist in sAD models and highlight the potential role of Aβ clearance mechanisms, a relatively under-investigated area in iPSC-derived models, in the study of AD. We discuss the development of more physiologically relevant models using co-culture and three-dimensional culture of iPSC-derived neurons with glial cells. Finally, we evaluate whether we can develop better, more consistent models for sAD research using genetic stratification of iPSCs and identification of genetic and environmental risk factors that could be used to initiate disease onset for modelling sAD. These considerations provide exciting opportunities to develop more relevant iPSC models of sAD which can help drive our understanding of disease mechanisms and identify new therapeutic targets.
Collapse
Affiliation(s)
- Helen A Rowland
- Division of Neuroscience & Experimental Psychology, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK
| | - Nigel M Hooper
- Division of Neuroscience & Experimental Psychology, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK
| | - Katherine A B Kellett
- Division of Neuroscience & Experimental Psychology, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK.
| |
Collapse
|
19
|
Shackleton B, Crawford F, Bachmeier C. Apolipoprotein E-mediated Modulation of ADAM10 in Alzheimer's Disease. Curr Alzheimer Res 2018; 14:578-585. [PMID: 28164773 DOI: 10.2174/1567205014666170203093219] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 01/19/2017] [Accepted: 01/31/2017] [Indexed: 11/22/2022]
Abstract
BACKGROUND The APOE4 allele is the strongest genetic risk factor for Alzheimer's disease (AD). It has been associated with an accumulation of amyloid-β (Aβ) in the brain, which is produced through the sequential cleavage of the amyloid-β precursor protein (AβPP) by β - and γ-secretases. Alternatively, AβPP is also cleaved by α -secretases such as A Disintegrin and Metalloproteinase Domain-containing Protein 10 (ADAM10). OBJECTIVE While several studies have investigated the impact of apoE on β- and γ-secretase, interactions between apoE and α-secretases have not been fully examined. We investigated the effect of each apoE isoform on ADAM10 in vitro and in human cortex samples. METHOD ADAM10 activity and kinetics was assessed in cell-free assays and the biological activity of ADAM10 further investigated in 7WCHO cells over-expressing wild type AβPP through ELISA. Finally, ADAM10 expression and activity was observed in the soluble fraction of both control and Alzheimer's Disease human cortex samples through ELISA. RESULTS In a cell free assay, ADAM10 activity was found to be significantly lower in apoE4 samples compared to apoE2. 7WCHO cells over expressing wild type AβPP exposed to apoE4 demonstrated reduced formation of sAβPPα compared to other apoE isoforms. We also identified APOE and AD dependent changes in ADAM10 activity and expression in the soluble brain fraction of human brain cortex. CONCLUSION Overall, our data demonstrates an apoE isoform-dependent effect on ADAM10 function and AβPP processing which may describe the elevated amyloid levels in the brains of AD subjects carrying the APOE4 allele.
Collapse
Affiliation(s)
- Ben Shackleton
- 2040 Whitfield Avenue, Sarasota, Florida 34243, United States
| | - Fiona Crawford
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, Florida, 34243, United States
| | - Corbin Bachmeier
- The Open University, Walton Hall, Milton Keynes, Buckinghamshire, MK7 6AA, United Kingdom
| |
Collapse
|
20
|
Inflammation as a Possible Link Between Dyslipidemia and Alzheimer’s Disease. Neuroscience 2018; 376:127-141. [PMID: 29454102 DOI: 10.1016/j.neuroscience.2018.02.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 02/05/2018] [Accepted: 02/07/2018] [Indexed: 01/08/2023]
|
21
|
Merino-Zamorano C, Fernández-de Retana S, Montañola A, Batlle A, Saint-Pol J, Mysiorek C, Gosselet F, Montaner J, Hernández-Guillamon M. Modulation of Amyloid-β1-40 Transport by ApoA1 and ApoJ Across an in vitro Model of the Blood-Brain Barrier. J Alzheimers Dis 2018; 53:677-91. [PMID: 27232214 DOI: 10.3233/jad-150976] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Amyloid-β (Aβ) accumulation in Alzheimer's disease (AD) and cerebral amyloid angiopathy (CAA) is likely caused by the impairment of its brain clearance that partly occurs through the blood-brain barrier (BBB). In this context, an in vitro BBB model is a valuable tool for studying the molecular mechanisms that regulate this process. This study assessed brain Aβ elimination across the BBB and its modulation by the natural chaperones Apolipoprotein A1 (ApoA1) and Apolipoprotein J/Clusterin (ApoJ). The model was based on primary cerebral endothelial cells that were cultured on Matrigel-coated Transwells and treated with fluorescently labeled-Aβ1-40 to track its efflux across the BBB, which corresponds to trafficking from the basolateral (brain) to apical (blood) compartments. We observed that the transport of basolateral Aβ1-40 was enhanced when it was complexed to rApoJ, whereas the complex formed with rApoA1 did not influence Aβ1-40 efflux. However, the presence of rApoA1 in the apical compartment was able to mobilize Aβ1-40 from the basolateral side. We also observed that both rApoA1 and rApoJ moderately crossed the monolayer (from blood to brain) through a mechanism involving the LDL receptor-related protein family. In contrast to the increased rApoJ efflux when complexed to Aβ1-40, rApoA1 trafficking was restricted when it was bound to the Aβ peptide. In summary, the present study highlights the role of ApoJ and ApoA1 in the in vitro modulation of Aβ elimination across the BBB.
Collapse
Affiliation(s)
- Cristina Merino-Zamorano
- Neurovascular Research Laboratory, Vall d'Hebron Research Insitute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Sofía Fernández-de Retana
- Neurovascular Research Laboratory, Vall d'Hebron Research Insitute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alex Montañola
- Neurovascular Research Laboratory, Vall d'Hebron Research Insitute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Aina Batlle
- Neurovascular Research Laboratory, Vall d'Hebron Research Insitute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Julien Saint-Pol
- Univ. Artois, EA2465, Laboratoire de la Barrière Hémato-Encéphalique, LBHE, Lens, F-62300, France
| | - Caroline Mysiorek
- Univ. Artois, EA2465, Laboratoire de la Barrière Hémato-Encéphalique, LBHE, Lens, F-62300, France
| | - Fabien Gosselet
- Univ. Artois, EA2465, Laboratoire de la Barrière Hémato-Encéphalique, LBHE, Lens, F-62300, France
| | - Joan Montaner
- Neurovascular Research Laboratory, Vall d'Hebron Research Insitute, Universitat Autònoma de Barcelona, Barcelona, Spain.,Department of Neurology, Neurovascular Unit, Vall d'Hebron Hospital, Barcelona, Spain
| | - Mar Hernández-Guillamon
- Neurovascular Research Laboratory, Vall d'Hebron Research Insitute, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
22
|
McInerney MP, Short JL, Nicolazzo JA. Neurovascular Alterations in Alzheimer's Disease: Transporter Expression Profiles and CNS Drug Access. AAPS JOURNAL 2017; 19:940-956. [PMID: 28462473 DOI: 10.1208/s12248-017-0077-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 03/15/2017] [Indexed: 01/05/2023]
Abstract
Despite a century of steady and incremental progress toward understanding the underlying biochemical mechanisms, Alzheimer's disease (AD) remains a complicated and enigmatic disease, and greater insight will be necessary before substantive clinical success is realised. Over the last decade in particular, a large body of work has highlighted the cerebral microvasculature as an anatomical region with an increasingly apparent role in the pathogenesis of AD. The causative interplay and temporal cascade that manifest between the brain vasculature and the wider disease progression of AD are not yet fully understood, and further inquiry is required to properly characterise these relationships. The purpose of this review is to highlight the recent advancements in research implicating neurovascular factors in AD, at both the molecular and anatomical levels. We begin with a brief introduction of the biochemical and genetic aspects of AD, before reviewing the essential concepts of the blood-brain barrier (BBB) and the neurovascular unit (NVU). In detail, we then examine the evidence demonstrating involvement of BBB dysfunction in AD pathogenesis, highlighting the importance of neurovascular components in AD. Lastly, we include within this review research that focuses on how altered properties of the BBB in AD impact upon CNS exposure of therapeutic agents and the potential clinical impact that this may have on people with this disease.
Collapse
Affiliation(s)
- Mitchell P McInerney
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia
| | - Jennifer L Short
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, 3052, VIC, Australia
| | - Joseph A Nicolazzo
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia.
| |
Collapse
|
23
|
The Complex Role of Apolipoprotein E in Alzheimer's Disease: an Overview and Update. J Mol Neurosci 2016; 60:325-335. [PMID: 27647307 DOI: 10.1007/s12031-016-0839-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 09/05/2016] [Indexed: 10/21/2022]
Abstract
Apolipoprotein E (ApoE) plays a crucial role in the homeostatic control of lipids in both the periphery and the central nervous system (CNS). In humans, ApoE exists in three different isoforms: ε2, ε3 and ε4. ApoE ε3 is the most common isoform, while the ε4 isoform confers the greatest genetic risk for Alzheimer's disease (AD). However, the mechanisms underlying how ApoE contributes to the pathogenesis of AD are still debated. ApoE has been shown to impact amyloid β (Aβ) deposition and clearance in the brain. ApoE also has Aβ-independent pathways in AD, which has led to the discovery of new roles of ApoE ranging from mitochondria dysfunction to, most recently, iron metabolism. Here, we review the role of ApoE in health and in AD, with the view of identifying therapeutic approaches that could prevent the risk associated with the ε4 isoform.
Collapse
|
24
|
Shackleton B, Crawford F, Bachmeier C. Inhibition of ADAM10 promotes the clearance of Aβ across the BBB by reducing LRP1 ectodomain shedding. Fluids Barriers CNS 2016; 13:14. [PMID: 27503326 PMCID: PMC4977753 DOI: 10.1186/s12987-016-0038-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 08/01/2016] [Indexed: 12/22/2022] Open
Abstract
Background Transport across the blood–brain barrier (BBB) is an important mediator of beta-amyloid (Aβ) accumulation in the brain and a contributing factor in the pathogenesis of Alzheimer’s disease (AD). One of the receptors responsible for the transport of Aβ in the BBB is the low density lipoprotein receptor-related protein 1 (LRP1). LRP1 is susceptible to proteolytic shedding at the cell surface, which prevents endocytic transport of ligands. Previously, we reported a strong inverse correlation between LRP1 shedding in the brain and Aβ transit across the BBB. Several proteases contribute to the ectodomain shedding of LRP1 including the α-secretase, a desintegrin and metalloproteinase domain containing protein 10 (ADAM10). Methods The role of ADAM10 in the shedding of LRP1 and Aβ BBB clearance was assessed through pharmacological inhibition of ADAM10 in an in vitro model of the BBB and through the use of ADAM10 endothelial specific knock-out mice. In addition, an acute treatment paradigm with an ADAM10 inhibitor was also tested in an AD mouse model to assess the effect of ADAM10 inhibition on LRP1 shedding and Aβbrain accumulation. Results In the current studies, inhibition of ADAM10 reduced LRP1 shedding in brain endothelial cultures and increased Aβ42 transit across an in vitro model of the BBB. Similarly, transgenic ADAM10 endothelial knockout mice displayed lower LRP1 shedding in the brain and significantly enhanced Aβ clearance across the BBB compared to wild-type animals. Acute treatment with the ADAM10-selective inhibitor GI254023X in an AD mouse model substantially reduced brain LRP1 shedding and increased Aβ40 levels in the plasma, indicating enhanced Aβ transit from the brain to the periphery. Furthermore, both soluble and insoluble Aβ40 and Aβ42 brain levels were decreased following GI254023X treatment, but these effects lacked statistical significance. Conclusions These studies demonstrate a role for ADAM10 in the ectodomain shedding of LRP1 in the brain and the clearance of Aβ across the BBB, which may provide a novel strategy for attenuating Aβ accumulation in the AD brain.
Collapse
Affiliation(s)
- B Shackleton
- The Roskamp Institute, Sarasota, FL, 34243, USA. .,The Open University, Milton Keynes, Buckinghamshire, MK7 6AA, UK.
| | - F Crawford
- The Roskamp Institute, Sarasota, FL, 34243, USA.,The Open University, Milton Keynes, Buckinghamshire, MK7 6AA, UK
| | - C Bachmeier
- The Roskamp Institute, Sarasota, FL, 34243, USA.,The Open University, Milton Keynes, Buckinghamshire, MK7 6AA, UK
| |
Collapse
|
25
|
Kim HJ, Park KW, Kim TE, Im JY, Shin HS, Kim S, Lee DH, Ye BS, Kim JH, Kim EJ, Park KH, Han HJ, Jeong JH, Choi SH, Park SA. Elevation of the Plasma Aβ40/Aβ42 Ratio as a Diagnostic Marker of Sporadic Early-Onset Alzheimer's Disease. J Alzheimers Dis 2016; 48:1043-50. [PMID: 26444752 DOI: 10.3233/jad-143018] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Although plasma amyloid-β (Aβ) levels have been evaluated as a possible diagnostic marker of Alzheimer's disease (AD), the findings are inconsistent. OBJECTIVE The present study aimed to validate plasma levels of Aβ40, Aβ42, and the Aβ40/Aβ42 ratio as biomarkers of AD in subjects with early-onset AD (EOAD) without familial AD genetic mutations. METHODS Patients with sporadic EOAD (sEOAD) were prospectively recruited by nine neurology clinics. Plasma levels of Aβ40 and Aβ42 were measured using a sandwich enzyme-linked immunosorbent assay (ELISA) in 100 sEOAD (50-69 year-old) and 46 age-matched normal control subjects (50-72 year-old). Cerebrospinal fluid (CSF) was obtained from 32 sEOAD subjects and 25 controls. The integrity of the blood-brain barrier was assessed using the CSF/plasma albumin ratio. RESULTS The plasma levels of Aβ42 were significantly lower, while the Aβ40/Aβ42 ratio was significantly higher in sEOAD patients than in controls. The levels of Aβ40, Aβ42, and the Aβ40/Aβ42 ratio did not differ in relation to the APOEɛ4 allele. The CSF/plasma albumin ratio was comparable between the two groups, and the plasma parameters of Aβ proteins were not significantly associated. A multivariate analysis revealed that an increased Aβ40/Aβ42 ratio is valuable for the discrimination of sEOAD from controls (β=0.344, p=0.000). The area under the ROC curve for the Aβ40/Aβ42 ratio was 0.76, and a cut-off ratio of 5.87 was suggested to have 70% sensitivity and 68% specificity. CONCLUSION The plasma Aβ40/Aβ42 ratio had moderate validity for the discrimination of sEOAD patients from age-matched controls.
Collapse
Affiliation(s)
- Hyeong Jun Kim
- Department of Neurology, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Kyung Won Park
- Department of Neurology, Dong-A University College of Medicine, Dong-A University Hospital, Busan, Korea
| | - Tae Eun Kim
- Department of Neurology, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Ji Young Im
- Department of Neurology, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Ho Sik Shin
- Department of Neurology, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Saeromi Kim
- Department of Neurology, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Dong Hyun Lee
- Department of Neurology, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Byoung Seok Ye
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | - Jong Hun Kim
- Department of Neurology, Ilsan Hospital, National Health Insurance Corporation, Goyang, Korea
| | - Eun-Joo Kim
- Department of Neurology, Pusan National University Hospital, Busan, Korea
| | - Kee Hyung Park
- Department of Neurology, Gachon University, College of Medicine, Incheon, Korea
| | - Hyun Jeong Han
- Department of Neurology, Myongii Hospital, Seonam University College of Medicine, Goyang, Korea
| | - Jee Hyang Jeong
- Department of Neurology, Ewha Womans University Mokdong Hospital, Seoul, Korea
| | - Seong Hye Choi
- Department of Neurology, Inha University School of Medicine, Incheon, Korea
| | - Sun Ah Park
- Department of Neurology, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| |
Collapse
|
26
|
Tai LM, Thomas R, Marottoli FM, Koster KP, Kanekiyo T, Morris AWJ, Bu G. The role of APOE in cerebrovascular dysfunction. Acta Neuropathol 2016; 131:709-23. [PMID: 26884068 DOI: 10.1007/s00401-016-1547-z] [Citation(s) in RCA: 153] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 02/10/2016] [Accepted: 02/10/2016] [Indexed: 11/30/2022]
Abstract
The ε4 allele of the apolipoprotein E gene (APOE4) is associated with cognitive decline during aging, is the greatest genetic risk factor for Alzheimer's disease and has links to other neurodegenerative conditions that affect cognition. Increasing evidence indicates that APOE genotypes differentially modulate the function of the cerebrovasculature (CV), with apoE and its receptors expressed by different cell types at the CV interface (astrocytes, pericytes, smooth muscle cells, brain endothelial cells). However, research on the role of apoE in CV dysfunction has not advanced as quickly as other apoE-modulated pathways. This review will assess what aspects of the CV are modulated by APOE genotypes during aging and under disease states, discuss potential mechanisms, and summarize the therapeutic significance of the topic. We propose that APOE4 induces CV dysfunction through direct signaling at the CV, and indirectly via modulation of peripheral and central pathways. Further, that APOE4 predisposes the CV to damage by, and exacerbates the effects of, additional risk factors (such as sex, hypertension, and diabetes). ApoE4-induced detrimental CV changes include reduced cerebral blood flow (CBF), modified neuron-CBF coupling, increased blood-brain barrier leakiness, cerebral amyloid angiopathy, hemorrhages and disrupted transport of nutrients and toxins. The apoE4-induced detrimental changes may be linked to pericyte migration/activation, astrocyte activation, smooth muscle cell damage, basement membrane degradation and alterations in brain endothelial cells.
Collapse
Affiliation(s)
- Leon M Tai
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, 808 S.Wood St., M/C 512, Chicago, IL, 60612, USA.
| | - Riya Thomas
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, 808 S.Wood St., M/C 512, Chicago, IL, 60612, USA
| | - Felecia M Marottoli
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, 808 S.Wood St., M/C 512, Chicago, IL, 60612, USA
| | - Kevin P Koster
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, 808 S.Wood St., M/C 512, Chicago, IL, 60612, USA
| | - Takahisa Kanekiyo
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Alan W J Morris
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, 808 S.Wood St., M/C 512, Chicago, IL, 60612, USA
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| |
Collapse
|
27
|
Anderson-Mooney AJ, Schmitt FA, Head E, Lott IT, Heilman KM. Gait dyspraxia as a clinical marker of cognitive decline in Down syndrome: A review of theory and proposed mechanisms. Brain Cogn 2016; 104:48-57. [PMID: 26930369 PMCID: PMC4801771 DOI: 10.1016/j.bandc.2016.02.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 02/13/2016] [Accepted: 02/21/2016] [Indexed: 12/15/2022]
Abstract
Down syndrome (DS) is the most common genetic cause of intellectual disability in children. With aging, DS is associated with an increased risk for Alzheimer's disease (AD). The development of AD neuropathology in individuals with DS can result in further disturbances in cognition and behavior and may significantly exacerbate caregiver burden. Early detection may allow for appropriate preparation by caregivers. Recent literature suggests that declines in gait may serve as an early marker of AD-related cognitive disorders; however, this relationship has not been examined in individuals with DS. The theory regarding gait dyspraxia and cognitive decline in the general population is reviewed, and potential applications to the population with individuals with DS are highlighted. Challenges and benefits in the line of inquiry are discussed. In particular, it appears that gait declines in aging individuals with DS may be associated with known declines in frontoparietal gray matter, development of AD-related pathology, and white matter losses in tracts critical to motor control. These changes are also potentially related to the cognitive and functional changes often observed during the same chronological period as gait declines in adults with DS. Gait declines may be an early marker of cognitive change, related to the development of underlying AD-related pathology, in individuals with DS. Future investigations in this area may provide insight into the clinical changes associated with development of AD pathology in both the population with DS and the general population, enhancing efforts for optimal patient and caregiver support and propelling investigations regarding safety/quality of life interventions and disease-modifying interventions.
Collapse
Affiliation(s)
- Amelia J Anderson-Mooney
- University of Kentucky College of Medicine, Department of Neurology, 740 S. Limestone, Suite B-101, Lexington, KY 40536, United States.
| | - Frederick A Schmitt
- University of Kentucky College of Medicine, Department of Neurology and Sanders-Brown Center on Aging, 800 S. Limestone, Room 312, Lexington, KY 40536, United States.
| | - Elizabeth Head
- University of Kentucky, Department of Molecular & Biomedical Pharmacology and Sanders-Brown Center on Aging, 800 S. Limestone, Room 203, Lexington, KY 40536, United States.
| | - Ira T Lott
- University of California - Irvine School of Medicine, Department of Pediatrics, Bldg 2 3rd Floor Rt 81, 101 The City Drive, Mail Code: 4482, Orange, CA 92668, United States.
| | - Kenneth M Heilman
- University of Florida College of Medicine, Department of Neurology, Room L3-100, McKnight Brain Institute, 1149 Newell Drive, Gainesville, FL 32611, United States.
| |
Collapse
|
28
|
Lu J, Fu J, Zhong Y, Chen P, Yang Q, Zhao Y, Wan L, Guo C. The roles of apolipoprotein E3 and CYP2D6 (rs1065852) gene polymorphisms in the predictability of responses to individualized therapy with donepezil in Han Chinese patients with Alzheimer's disease. Neurosci Lett 2016; 614:43-8. [DOI: 10.1016/j.neulet.2015.12.062] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 12/30/2015] [Accepted: 12/30/2015] [Indexed: 11/26/2022]
|
29
|
Kyrtsos CR, Baras JS. Modeling the Role of the Glymphatic Pathway and Cerebral Blood Vessel Properties in Alzheimer's Disease Pathogenesis. PLoS One 2015; 10:e0139574. [PMID: 26448331 PMCID: PMC4598011 DOI: 10.1371/journal.pone.0139574] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 09/15/2015] [Indexed: 11/18/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common cause of dementia in the elderly, affecting over 10% population over the age of 65 years. Clinically, AD is described by the symptom set of short term memory loss and cognitive decline, changes in mentation and behavior, and eventually long-term memory deficit as the disease progresses. On imaging studies, significant atrophy with subsequent increase in ventricular volume have been observed. Pathology on post-mortem brain specimens demonstrates the classic findings of increased beta amyloid (Aβ) deposition and the presence of neurofibrillary tangles (NFTs) within affected neurons. Neuroinflammation, dysregulation of blood-brain barrier transport and clearance, deposition of Aβ in cerebral blood vessels, vascular risk factors such as atherosclerosis and diabetes, and the presence of the apolipoprotein E4 allele have all been identified as playing possible roles in AD pathogenesis. Recent research has demonstrated the importance of the glymphatic system in the clearance of Aβ from the brain via the perivascular space surrounding cerebral blood vessels. Given the variety of hypotheses that have been proposed for AD pathogenesis, an interconnected, multilayer model offers a unique opportunity to combine these ideas into a single unifying model. Results of this model demonstrate the importance of vessel stiffness and heart rate in maintaining adequate clearance of Aβ from the brain.
Collapse
Affiliation(s)
- Christina Rose Kyrtsos
- University of Pittsburgh Medical Center, Department of Neurology, Pittsburgh, Pennsylvania, United States of America
- Institute for Systems Research, University of Maryland, College Park, Maryland, United States of America
| | - John S. Baras
- Institute for Systems Research, University of Maryland, College Park, Maryland, United States of America
- Department of Electrical and Computer Engineering, University of Maryland, College Park, Maryland, United States of America
- * E-mail:
| |
Collapse
|
30
|
Liu DS, Pan XD, Zhang J, Shen H, Collins NC, Cole AM, Koster KP, Ben Aissa M, Dai XM, Zhou M, Tai LM, Zhu YG, LaDu M, Chen XC. APOE4 enhances age-dependent decline in cognitive function by down-regulating an NMDA receptor pathway in EFAD-Tg mice. Mol Neurodegener 2015; 10:7. [PMID: 25871877 PMCID: PMC4391134 DOI: 10.1186/s13024-015-0002-2] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 01/26/2015] [Indexed: 12/16/2022] Open
Abstract
Background Alzheimer’s disease (AD) causes progressive loss of memory and cognition, exacerbated by APOE4, the greatest genetic risk factor for AD. One proposed mechanism for apolipoprotein E (apoE) effects on cognition is via NMDAR-dependent signaling. APOE genotype-specific effects on this pathway were dissected using EFAD-transgenic (Tg) mice (5xFAD mice, that over-express human amyloid-beta (Aβ) via 5 familial-AD (FAD) mutations, and express human apoE), and 5xFAD/APOE-knockout (KO) mice. Previous data from EFAD-Tg mice demonstrate age-dependent (2-6 months), apoE-specific effects on the development of Aβ pathology. This study tests the hypothesis that apoE4 impairs cognition via modulation of NMDAR-dependent signaling, specifically via a loss of function by comparison of E4FAD mice with 5xFAD/APOE-KO mice, E3FAD and E2FAD mice. Results Using female E2FAD, E3FAD, E4FAD and 5xFAD/APOE-KO mice aged 2-, 4-, and 6-months, the Y-maze and Morris water maze behavioral tests were combined with synaptic protein levels as markers of synaptic viability. The results demonstrate a greater age-induced deficit in cognition and reduction in PSD95, drebrin and NMDAR subunits in the E4FAD and 5xFAD/APOE-KO mice compared with E2FAD and E3FAD mice, consistent with an apoE4 loss of function. Interestingly, for NMDAR-mediated signaling, the levels of p-CaMK-II followed this same apoE-specific pattern as cognition, while the levels of p-CREB and BDNF demonstrate an apoE4 toxic gain of function: E2FAD > E3FAD > 5xFAD/APOE-KO > E4FAD. Conclusion These findings suggest that compared with E2FAD and E3FAD, E4FAD and 5xFAD/APOE-KO mice exhibit enhanced age-induced reductions in cognition and key synaptic proteins via down-regulation of an NMDAR signaling pathway, consistent with an apoE4 loss of function. However, levels of p-CREB and BDNF, signaling factors common to multiple pathways, suggest a gain of toxic function. Publications in this field present contradictory results as to whether APOE4 imparts a loss or gain of function. As with the results reported herein, the overall effect of APOE4 on a given CNS-specific measure will be the product of multiple overlapping mechanisms. Thus, caution remains critical in determining whether APOE gene inactivation or therapies that correct the loss of positive function related to apoE4, are the appropriate therapeutic response. Electronic supplementary material The online version of this article (doi:10.1186/s13024-015-0002-2) contains supplementary material, which is available to authorized users.
Collapse
|
31
|
Esiri M, Chance S, Joachim C, Warden D, Smallwood A, Sloan C, Christie S, Wilcock G, Smith AD. Cerebral amyloid angiopathy, subcortical white matter disease and dementia: literature review and study in OPTIMA. Brain Pathol 2015; 25:51-62. [PMID: 25521177 PMCID: PMC8028928 DOI: 10.1111/bpa.12221] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 10/08/2014] [Indexed: 12/23/2022] Open
Abstract
Cerebral amyloid angiopathy (CAA) is of increasing clinical and research interest as the ability to detect it and its consequences by neuroimaging in living subjects has advanced. There is also increasing interest in understanding its possible role in the development of intracerebral hemorrhage, Alzheimer's disease (AD) and vascular dementia. In this article, the literature on this subject is reviewed and novel findings relating CAA to subcortical white matter damage in 224 subjects in the Oxford project to Investigate Memory and Ageing (OPTIMA) are reported. The relationship between CAA and subcortical tissue damage in the OPTIMA subjects was found to be critically dependent on ApoE genotype, there being a positive relationship between measures of CAA and subcortical small vessel disease in ApoEε4 carriers and a significant negative relationship in ApoEε2 carriers. These findings draw attention, as have many other studies, to the importance of ApoE genotype as a major risk factor not only for dementia but also for damage to blood vessels in the aging brain.
Collapse
Affiliation(s)
- Margaret Esiri
- Neuropathology DepartmentNuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | - Steven Chance
- Neuropathology DepartmentNuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | - Catharine Joachim
- Neuropathology DepartmentNuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | - Donald Warden
- Department of PharmacologyUniversity of OxfordOxfordUK
| | | | - Carolyn Sloan
- Neuropathology DepartmentNuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | - Sharon Christie
- Nuffield Department of Clinical NeurosciencesOPTIMAUniversity of OxfordOxfordUK
| | - Gordon Wilcock
- Nuffield Department of Clinical NeurosciencesOPTIMAUniversity of OxfordOxfordUK
| | | |
Collapse
|
32
|
Bachmeier C, Shackleton B, Ojo J, Paris D, Mullan M, Crawford F. Apolipoprotein E isoform-specific effects on lipoprotein receptor processing. Neuromolecular Med 2014; 16:686-96. [PMID: 25015123 PMCID: PMC4280344 DOI: 10.1007/s12017-014-8318-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 07/01/2014] [Indexed: 01/24/2023]
Abstract
Recent findings indicate an isoform-specific role for apolipoprotein E (apoE) in the elimination of beta-amyloid (Aβ) from the brain. ApoE is closely associated with various lipoprotein receptors, which contribute to Aβ brain removal via metabolic clearance or transit across the blood–brain barrier (BBB). These receptors are subject to ectodomain shedding at the cell surface, which alters endocytic transport and mitigates Aβ elimination. To further understand the manner in which apoE influences Aβ brain clearance, these studies investigated the effect of apoE on lipoprotein receptor shedding. Consistent with prior reports, we observed an increased shedding of the low-density lipoprotein receptor (LDLR) and the LDLR-related protein 1 (LRP1) following Aβ exposure in human brain endothelial cells. When Aβ was co-treated with each apoE isoform, there was a reduction in Aβ-induced shedding with apoE2 and apoE3, while lipoprotein receptor shedding in the presence of apoE4 remained increased. Likewise, intracranial administration of Aβ to apoE-targeted replacement mice (expressing the human apoE isoforms) resulted in an isoform-dependent effect on lipoprotein receptor shedding in the brain (apoE4 > apoE3 > apoE2). Moreover, these results show a strong inverse correlation with our prior work in apoE transgenic mice in which apoE4 animals showed reduced Aβ clearance across the BBB compared to apoE3 animals. Based on these results, apoE4 appears less efficient than other apoE isoforms in regulating lipoprotein receptor shedding, which may explain the differential effects of these isoforms in removing Aβ from the brain.
Collapse
Affiliation(s)
- Corbin Bachmeier
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, Florida 34243, USA
| | - Ben Shackleton
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, Florida 34243, USA
| | - Joseph Ojo
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, Florida 34243, USA
| | - Daniel Paris
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, Florida 34243, USA
| | - Michael Mullan
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, Florida 34243, USA
| | - Fiona Crawford
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, Florida 34243, USA
| |
Collapse
|
33
|
Beta-amyloid auto-antibodies are reduced in Alzheimer's disease. J Neuroimmunol 2014; 274:168-73. [PMID: 25022335 DOI: 10.1016/j.jneuroim.2014.06.017] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Revised: 05/29/2014] [Accepted: 06/19/2014] [Indexed: 01/01/2023]
Abstract
Accumulation and cytotoxicity of amyloid beta (Aβ) are understood as the major cause of Alzheimer's disease (AD). There is evidence that naturally occurring antibodies against amyloid beta (Aβ) protein play a role in Aβ-clearance, and such a mechanism appears to be impaired in AD. In the present study, the anti-Aβ antibodies in the serum from individuals with and without late onset AD were measured using ELISA and dot-blot methods. Aβ auto-antibodies in serum were mainly targeted to Aβ1-15 epitope and its titer was significantly lower in AD patients than elderly non-AD controls (NC). The dot-blot analysis further demonstrated that auto-antibodies against fibrillar Aβ42, Aβ1-15 and Aβ16-30 epitopes were all in a lower level in AD than in NC. The isotypes of the auto-antibodies were mainly non-inflammatory IgG2 type. We also analyzed the relationship of auto-Aβ antibody levels with the genotypes of apolipoprotein E (ApoE) and ANKK1/DRD2 gene.
Collapse
|
34
|
Abstract
The vast majority of Alzheimer's disease (AD) cases are late onset (LOAD), which is genetically complex with heritability estimates up to 80%. Apolipoprotein E (APOE) has been irrefutably recognized as the major genetic risk factor, with semidominant inheritance, for LOAD. Although the mechanisms that underlie the pathogenic nature of APOE in AD are still not completely understood, emerging data suggest that APOE contributes to AD pathogenesis through both amyloid-β (Aβ)-dependent and Aβ-independent pathways. Given the central role for APOE in the modulation of AD pathogenesis, many therapeutic strategies have emerged, including converting APOE conformation, regulating APOE expression, mimicking APOE peptides, blocking the APOE/Aβ interaction, modulating APOE lipidation state, and gene therapy. Accumulating evidence also suggests the utility of APOE genotyping in AD diagnosis, risk assessment, prevention, and treatment response.
Collapse
Affiliation(s)
- Jin-Tai Yu
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao 266071, China; ,
| | | | | |
Collapse
|
35
|
Swaminathan S, Risacher SL, Yoder KK, West JD, Shen L, Kim S, Inlow M, Foroud T, Jagust WJ, Koeppe RA, Mathis CA, Shaw LM, Trojanowski JQ, Soares H, Aisen PS, Petersen RC, Weiner MW, Saykin AJ. Association of plasma and cortical amyloid beta is modulated by APOE ε4 status. Alzheimers Dement 2014; 10:e9-e18. [PMID: 23541187 PMCID: PMC3750076 DOI: 10.1016/j.jalz.2013.01.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Revised: 01/10/2013] [Accepted: 01/16/2013] [Indexed: 10/27/2022]
Abstract
BACKGROUND Apolipoprotein E (APOE) ε4 allele's role as a modulator of the relationship between soluble plasma amyloid beta (Aβ) and fibrillar brain Aβ measured by Pittsburgh compound B positron emission tomography ([(11)C]PiB PET) has not been assessed. METHODS Ninety-six Alzheimer's Disease Neuroimaging Initiative participants with [(11)C]PiB scans and plasma Aβ1-40 and Aβ1-42 measurements at the time of PET scanning were included. Regional and voxelwise analyses of [(11)C]PiB data were used to determine the influence of APOE ε4 allele on association of plasma Aβ1-40, Aβ1-42, and Aβ1-40/Aβ1-42 with [(11)C]PiB uptake. RESULTS In APOE ε4- but not ε4+ participants, positive relationships between plasma Aβ1-40/Aβ1-42 and [(11)C]PiB uptake were observed. Modeling the interaction of APOE and plasma Aβ1-40/Aβ1-42 improved the explained variance in [(11)C]PiB binding compared with using APOE and plasma Aβ1-40/Aβ1-42 as separate terms. CONCLUSIONS The results suggest that plasma Aβ is a potential Alzheimer's disease biomarker and highlight the importance of genetic variation in interpretation of plasma Aβ levels.
Collapse
Affiliation(s)
- Shanker Swaminathan
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Shannon L Risacher
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Karmen K Yoder
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - John D West
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Li Shen
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sungeun Kim
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Mark Inlow
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Mathematics, Rose-Hulman Institute of Technology, Terre Haute, IN, USA
| | - Tatiana Foroud
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA; Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - William J Jagust
- Helen Wills Neuroscience Institute, University of California at Berkeley, Berkeley, CA, USA
| | - Robert A Koeppe
- Division of Nuclear Medicine, Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | - Chester A Mathis
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Leslie M Shaw
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | | | - Paul S Aisen
- Department of Neurosciences, University of California at San Diego, San Diego, CA, USA
| | - Ronald C Petersen
- Department of Neurology, Mayo Clinic and Foundation, Rochester, MN, USA
| | - Michael W Weiner
- Department of Veterans Affairs Medical Center, Center for Imaging of Neurodegenerative Diseases, San Francisco, CA, USA
| | - Andrew J Saykin
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA; Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
36
|
Erickson MA, Banks WA. Blood-brain barrier dysfunction as a cause and consequence of Alzheimer's disease. J Cereb Blood Flow Metab 2013; 33:1500-13. [PMID: 23921899 PMCID: PMC3790938 DOI: 10.1038/jcbfm.2013.135] [Citation(s) in RCA: 414] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Revised: 07/05/2013] [Accepted: 07/09/2013] [Indexed: 12/14/2022]
Abstract
The blood-brain barrier (BBB) plays critical roles in the maintenance of central nervous system (CNS) homeostasis. Dysfunction of the BBB occurs in a number of CNS diseases, including Alzheimer's disease (AD). A prevailing hypothesis in the AD field is the amyloid cascade hypothesis that states that amyloid-β (Aβ) deposition in the CNS initiates a cascade of molecular events that cause neurodegeneration, leading to AD onset and progression. In this review, the participation of the BBB in the amyloid cascade and in other mechanisms of AD neurodegeneration will be discussed. We will specifically focus on three aspects of BBB dysfunction: disruption, perturbation of transporters, and secretion of neurotoxic substances by the BBB. We will also discuss the interaction of the BBB with components of the neurovascular unit in relation to AD and the potential contribution of AD risk factors to aspects of BBB dysfunction. From the results discussed herein, we conclude that BBB dysfunction contributes to AD through a number of mechanisms that could be initiated in the presence or absence of Aβ pathology.
Collapse
Affiliation(s)
- Michelle A Erickson
- 1] GRECC, Veterans Affairs Puget Sound Health Care System, Seattle, Washington, USA [2] Division of Gerontology and Geriatric Medicine, Department of Internal Medicine, University of Washington School of Medicine, Seattle, Washington, USA [3] Department of Pathology, School of Dental Medicine, University of Pennsylvania, Seattle, Washington, USA
| | | |
Collapse
|
37
|
Bachmeier C, Beaulieu-Abdelahad D, Mullan M, Paris D. Role of the cannabinoid system in the transit of beta-amyloid across the blood-brain barrier. Mol Cell Neurosci 2013; 56:255-62. [PMID: 23831388 DOI: 10.1016/j.mcn.2013.06.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Revised: 05/30/2013] [Accepted: 06/25/2013] [Indexed: 12/01/2022] Open
Abstract
Emerging evidence suggests beta-amyloid (Aβ) deposition in the Alzheimer's disease (AD) brain is the result of impaired clearance, due in part to diminished Aβ transport across the blood-brain barrier (BBB). Recently, modulation of the cannabinoid system was shown to reduce Aβ brain levels and improve cognitive behavior in AD animal models. The purpose of the current studies was to investigate the role of the cannabinoid system in the clearance of Aβ across the BBB. Using in vitro and in vivo models of BBB clearance, Aβ transit across the BBB was examined in the presence of cannabinoid receptor agonists and inhibitors. In addition, expression levels of the Aβ transport protein, lipoprotein receptor-related protein1 (LRP1), were determined in the brain and plasma of mice following cannabinoid treatment. Cannabinoid receptor agonism or inhibition of endocannabinoid-degrading enzymes significantly enhanced Aβ clearance across the BBB (2-fold). Moreover, cannabinoid receptor inhibition negated the stimulatory influence of cannabinoid treatment on Aβ BBB clearance. Additionally, LRP1 levels in the brain and plasma were elevated following cannabinoid treatment (1.5-fold), providing rationale for the observed increase in Aβ transit from the brain to the periphery. The current studies demonstrate, for the first time, a role for the cannabinoid system in the transit of Aβ across the BBB. These findings provide insight into the mechanism by which cannabinoid treatment reduces Aβ burden in the AD brain and offer additional evidence on the utility of this pathway as a treatment for AD.
Collapse
Affiliation(s)
- Corbin Bachmeier
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL 34243, USA.
| | | | | | | |
Collapse
|
38
|
Martiskainen H, Haapasalo A, Kurkinen KMA, Pihlajamäki J, Soininen H, Hiltunen M. Targeting ApoE4/ApoE receptor LRP1 in Alzheimer's disease. Expert Opin Ther Targets 2013; 17:781-94. [PMID: 23573918 DOI: 10.1517/14728222.2013.789862] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Progressive neuronal loss is a key feature in Alzheimer's disease (AD), which is the most common neurodegenerative disorder in the aging population. Currently, there are no therapeutic means to intervene neuronal damage in AD and therefore innovative approaches to discover novel strategies for the treatment of AD are needed. Based on the prevailing amyloid cascade hypothesis, it is conceivable that lowering the β-amyloid (Aβ) levels is sufficient to slow down the disease process, if started early enough. AREAS COVERED Here, we review genetic and biological functions related to apolipoprotein E (ApoE) and low-density lipoprotein receptor-related protein 1 receptor (LRP1)-mediated clearance of Aβ. Furthermore, we discuss the AD-related therapeutic potential of targeting to ApoE receptor LRP1 at the blood-brain barrier (BBB) and in the periphery. EXPERT OPINION Due to the recent setbacks in the clinical trials targeting AD, it is instrumental to seek alternative therapeutic approaches, which aim to reduce the accumulation of Aβ in the brain tissue. As the ApoE/LRP1-mediated clearance of Aβ across the BBB is the key event in the regulation of Aβ transcytosis from brain to periphery, direct targeting of this protein entity at the BBB holds a great potential in the treatment of AD.
Collapse
Affiliation(s)
- Henna Martiskainen
- Kuopio University Hospital, Institute of Clinical Medicine-Neurology, University of Eastern Finland and Department of Neurology, Kuopio, Finland
| | | | | | | | | | | |
Collapse
|
39
|
Bachmeier C, Beaulieu-Abdelahad D, Crawford F, Mullan M, Paris D. Stimulation of the Retinoid X Receptor Facilitates Beta-Amyloid Clearance Across the Blood–Brain Barrier. J Mol Neurosci 2012; 49:270-6. [DOI: 10.1007/s12031-012-9866-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Accepted: 07/24/2012] [Indexed: 12/12/2022]
|