1
|
Blömeke L, Rehn F, Kraemer‐Schulien V, Kutzsche J, Pils M, Bujnicki T, Lewczuk P, Kornhuber J, Freiesleben SD, Schneider L, Preis L, Priller J, Spruth EJ, Altenstein S, Lohse A, Schneider A, Fliessbach K, Wiltfang J, Hansen N, Rostamzadeh A, Düzel E, Glanz W, Incesoy EI, Butryn M, Buerger K, Janowitz D, Ewers M, Perneczky R, Rauchmann B, Teipel S, Kilimann I, Goerss D, Laske C, Munk MH, Sanzenbacher C, Spottke A, Roy‐Kluth N, Heneka MT, Brosseron F, Wagner M, Wolfsgruber S, Kleineidam L, Stark M, Schmid M, Jessen F, Bannach O, Willbold D, Peters O. Aβ oligomers peak in early stages of Alzheimer's disease preceding tau pathology. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2024; 16:e12589. [PMID: 38666085 PMCID: PMC11044868 DOI: 10.1002/dad2.12589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 03/11/2024] [Accepted: 03/15/2024] [Indexed: 04/28/2024]
Abstract
INTRODUCTION Soluble amyloid beta (Aβ) oligomers have been suggested as initiating Aβ related neuropathologic change in Alzheimer's disease (AD) but their quantitative distribution and chronological sequence within the AD continuum remain unclear. METHODS A total of 526 participants in early clinical stages of AD and controls from a longitudinal cohort were neurobiologically classified for amyloid and tau pathology applying the AT(N) system. Aβ and tau oligomers in the quantified cerebrospinal fluid (CSF) were measured using surface-based fluorescence intensity distribution analysis (sFIDA) technology. RESULTS Across groups, highest Aβ oligomer levels were found in A+ with subjective cognitive decline and mild cognitive impairment. Aβ oligomers were significantly higher in A+T- compared to A-T- and A+T+. APOE ε4 allele carriers showed significantly higher Aβ oligomer levels. No differences in tau oligomers were detected. DISCUSSION The accumulation of Aβ oligomers in the CSF peaks early within the AD continuum, preceding tau pathology. Disease-modifying treatments targeting Aβ oligomers might have the highest therapeutic effect in these disease stages. Highlights Using surface-based fluorescence intensity distribution analysis (sFIDA) technology, we quantified Aβ oligomers in cerebrospinal fluid (CSF) samples of the DZNE-Longitudinal Cognitive Impairment and Dementia (DELCODE) cohortAβ oligomers were significantly elevated in mild cognitive impairment (MCI)Amyloid-positive subjects in the subjective cognitive decline (SCD) group increased compared to the amyloid-negative control groupInterestingly, levels of Aβ oligomers decrease at advanced stages of the disease (A+T+), which might be explained by altered clearing mechanisms.
Collapse
|
2
|
Safieh M, Liraz O, Ovadia M, Michaelson D. The Role of Impaired Receptor Trafficking in Mediating the Pathological Effects of APOE4 in Alzheimer's Disease. J Alzheimers Dis 2024; 97:753-775. [PMID: 38217595 PMCID: PMC10894586 DOI: 10.3233/jad-230514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/06/2023] [Indexed: 01/15/2024]
Abstract
BACKGROUND Apolipoprotein E4 (APOE4) is the most prevalent genetic risk factor of Alzheimer's disease. Several studies suggest that APOE4 binding to its receptors is associated with their internalization and accumulation in intracellular compartments. Importantly, this phenomenon also occurs with other, non-ApoE receptors. Based on these observations, we hypothesized that APOE4 pathological effects are mediated by impairment in the life cycle of distinct receptors (APOER2, LRP1, IR, VEGFR). OBJECTIVE To examine the effects of APOE genotype on receptors protein levels and compartmentalization. METHODS Primary mouse neurons were prepared from APOE3 or APOE4 targeted replacement mice, or APOE-KO mice. Specific receptors protein levels were evaluated in these neurons, utilizing immunofluorescent staining. Additionally, surface membrane protein levels of those receptors were assessed by cell surface biotinylation assay and ELISA. Receptors' colocalization with intracellular compartments was assessed by double staining and confocal microscopy, followed by colocalization analysis. Finally, LRP1 or APOER2 were knocked-down with CRISPR/Cas9 system to examine their role in mediating APOE4 effects on the receptors. RESULTS Our results revealed lower receptors' levels in APOE4, specifically on the membrane surface. Additionally, APOE4 affects the compartmentation of these receptors in two patterns: the first was observed with LRP1 and was associated with decreased receptor levels in numerous intracellular compartments. The second was obtained with the other receptors and was associated with their accumulation in early endosomes and their decrease in the late endosomes. CONCLUSIONS These results provide a unifying mechanism, in which APOE4 drives the down regulation of various receptors, which plays important roles in distinct APOE4 related pathological processes.
Collapse
Affiliation(s)
- Mirna Safieh
- Department of Neurobiology, Sagol School of Neurosciences, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Ramat Aviv, Tel Aviv, Israel
| | - Ori Liraz
- Department of Neurobiology, Sagol School of Neurosciences, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Ramat Aviv, Tel Aviv, Israel
| | - Maayan Ovadia
- Department of Neurobiology, Sagol School of Neurosciences, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Ramat Aviv, Tel Aviv, Israel
| | - Danny Michaelson
- Department of Neurobiology, Sagol School of Neurosciences, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Ramat Aviv, Tel Aviv, Israel
| |
Collapse
|
3
|
Gaetani L, Chiasserini D, Paolini Paoletti F, Bellomo G, Parnetti L. Required improvements for cerebrospinal fluid-based biomarker tests of Alzheimer's disease. Expert Rev Mol Diagn 2023; 23:1195-1207. [PMID: 37902844 DOI: 10.1080/14737159.2023.2276918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 10/25/2023] [Indexed: 11/01/2023]
Abstract
INTRODUCTION Cerebrospinal fluid (CSF) biomarkers represent a well-established tool for diagnosing Alzheimer's disease (AD), independently from the clinical stage, by reflecting the presence of brain amyloidosis (A+) and tauopathy (T+). In front of this important achievement, so far, (i) CSF AD biomarkers have not yet been adopted for routine clinical use in all Centers dedicated to AD, mainly due to inter-lab variation and lack of internationally accepted cutoff values; (ii) we do need to add other biomarkers more suitable to correlate with the clinical stage and disease monitoring; (iii) we also need to detect the co-presence of other 'non-AD' pathologies. AREAS COVERED Efforts to establish standardized cutoff values based on large-scale multi-center studies are discussed. The influence of aging and comorbidities on CSF biomarker levels is also analyzed, and possible solutions are presented, i.e. complementing the A/T/(N) system with markers of axonal damage and synaptic derangement. EXPERT OPINION The first, mandatory need is to reach common cutoff values and defined (automated) methodologies for CSF AD biomarkers. To properly select subjects deserving CSF analysis, blood tests might represent the first-line approach. In those subjects undergoing CSF analysis, multiple biomarkers, able to give a comprehensive and personalized pathophysiological/prognostic information, should be included.
Collapse
Affiliation(s)
- Lorenzo Gaetani
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Davide Chiasserini
- Section of Physiology and Biochemistry, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Giovanni Bellomo
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Lucilla Parnetti
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
4
|
Prasansuklab A, Sukjamnong S, Theerasri A, Hu VW, Sarachana T, Tencomnao T. Transcriptomic analysis of glutamate-induced HT22 neurotoxicity as a model for screening anti-Alzheimer's drugs. Sci Rep 2023; 13:7225. [PMID: 37142620 PMCID: PMC10160028 DOI: 10.1038/s41598-023-34183-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 04/25/2023] [Indexed: 05/06/2023] Open
Abstract
Glutamate-induced neurotoxicity in the HT22 mouse hippocampal neuronal cell line has been recognized as a valuable cell model for the study of neurotoxicity associated with neurodegenerative diseases including Alzheimer's disease (AD). However, the relevance of this cell model for AD pathogenesis and preclinical drug screening remains to be more elucidated. While there is increasing use of this cell model in a number of studies, relatively little is known about its underlying molecular signatures in relation to AD. Here, our RNA sequencing study provides the first transcriptomic and network analyses of HT22 cells following glutamate exposure. Several differentially expressed genes (DEGs) and their relationships specific to AD were identified. Additionally, the usefulness of this cell model as a drug screening system was assessed by determining the expression of those AD-associated DEGs in response to two medicinal plant extracts, Acanthus ebracteatus and Streblus asper, that have been previously shown to be protective in this cell model. In summary, the present study reports newly identified AD-specific molecular signatures in glutamate-injured HT22 cells, suggesting that this cell can be a valuable model system for the screening and evaluation of new anti-AD agents, particularly from natural products.
Collapse
Affiliation(s)
- Anchalee Prasansuklab
- Natural Products for Neuroprotection and Anti-ageing Research Unit, Chulalongkorn University, Bangkok, 10330, Thailand
- College of Public Health Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Suporn Sukjamnong
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
- SYstems Neuroscience of Autism and PSychiatric Disorders (SYNAPS) Research Unit, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Atsadang Theerasri
- Natural Products for Neuroprotection and Anti-ageing Research Unit, Chulalongkorn University, Bangkok, 10330, Thailand
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Valerie W Hu
- Department of Biochemistry and Molecular Medicine, The George Washington University School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Tewarit Sarachana
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
- SYstems Neuroscience of Autism and PSychiatric Disorders (SYNAPS) Research Unit, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Tewin Tencomnao
- Natural Products for Neuroprotection and Anti-ageing Research Unit, Chulalongkorn University, Bangkok, 10330, Thailand.
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
5
|
Valencia-Olvera AC, Maldonado Weng J, Christensen A, LaDu MJ, Pike CJ. Role of estrogen in women's Alzheimer's disease risk as modified by APOE. J Neuroendocrinol 2023; 35:e13209. [PMID: 36420620 PMCID: PMC10049970 DOI: 10.1111/jne.13209] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 09/29/2022] [Accepted: 10/13/2022] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) is characterized by numerous sexual dimorphisms that impact the development, progression, and probably the strategies to prevent and treat the most common form of dementia. In this review, we consider this topic from a female perspective with a specific focus on how women's vulnerability to the disease is affected by the individual and interactive effects of estrogens and apolipoprotein E (APOE) genotype. Importantly, APOE appears to modulate systemic and neural outcomes of both menopause and estrogen-based hormone therapy. In the brain, dementia risk is greater in APOE4 carriers, and the impacts of hormone therapy on cognitive decline and dementia risk vary according to both outcome measure and APOE genotype. Beyond the CNS, estrogen and APOE genotype affect vulnerability to menopause-associated bone loss, dyslipidemia and cardiovascular disease risk. An emerging concept that may link these relationships is the possibility that the effects of APOE in women interact with estrogen status by mechanisms that may include modulation of estrogen responsiveness. This review highlights the need to consider the key AD risk factors of advancing age in a sex-specific manner to optimize development of therapeutic approaches for AD, a view aligned with the principle of personalized medicine.
Collapse
Affiliation(s)
- AC Valencia-Olvera
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - J Maldonado Weng
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - A Christensen
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089 USA
| | - MJ LaDu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - CJ Pike
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089 USA
| |
Collapse
|
6
|
Singh V, Mishra VN, Thakur MK. Identification of Plasma Proteomic Biomarkers in Patients with Mild Cognitive Impairment. Indian J Clin Biochem 2023; 38:33-41. [PMID: 36684491 PMCID: PMC9852370 DOI: 10.1007/s12291-022-01023-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 01/06/2022] [Indexed: 01/25/2023]
Abstract
Plasma proteomic profiling may provide novel biomarkers for the identification of mild cognitive impairment (MCI). The early diagnosis of MCI still remains a challenging task due to its diverse origin. Currently, molecular approaches have been used to identify MCI diversified origin as its onset is governed by a variety of molecular changes. Therefore, we aimed to find out molecular alteration in plasma using proteomics in patients with MCI for early detection of prodromal Alzheimer's disease (AD). To achieve this, we performed two-dimensional (2-D) gel electrophoresis coupled with MALDI-TOF/MS, which is used to analyze the differentially expressed proteins. In our study, we found three significantly altered proteins. Out of three differentially expressed proteins, one was downregulated and two were upregulated in MCI individuals as compared to control. Further, In silico analysis showed that identified proteins are involved in pathways such as complement and coagulation cascades, platelet activation and AD. STRING interaction network analysis revealed that the majority of proteins including apolipoprotein E (APO-E) have a common association with Transthyretin (TTR) and fibrinogen chain beta (FGB) protein. This suggests that APO-E, TTR and FGB are the key proteins with which other proteins interact to exert other biological functions. Conclusively, these proteins showing differential expression in the plasma might be used as a potent signature in blood for the diagnosis of MCI individuals.
Collapse
Affiliation(s)
- Vineeta Singh
- Department of Neurology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, UP 221005 India
| | - Vijaya Nath Mishra
- Department of Neurology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, UP 221005 India
| | - Mahendra Kumar Thakur
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, UP 221005 India
| |
Collapse
|
7
|
Zieneldien T, Kim J, Sawmiller D, Cao C. The Immune System as a Therapeutic Target for Alzheimer’s Disease. Life (Basel) 2022; 12:life12091440. [PMID: 36143476 PMCID: PMC9506058 DOI: 10.3390/life12091440] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/11/2022] [Accepted: 09/14/2022] [Indexed: 11/25/2022] Open
Abstract
Alzheimer’s disease (AD) is a heterogeneous neurodegenerative disorder and is the most common cause of dementia. Furthermore, aging is considered the most critical risk factor for AD. However, despite the vast amount of research and resources allocated to the understanding and development of AD treatments, setbacks have been more prominent than successes. Recent studies have shown that there is an intricate connection between the immune and central nervous systems, which can be imbalanced and thereby mediate neuroinflammation and AD. Thus, this review examines this connection and how it can be altered with AD. Recent developments in active and passive immunotherapy for AD are also discussed as well as suggestions for improving these therapies moving forward.
Collapse
Affiliation(s)
- Tarek Zieneldien
- Department of Pharmaceutical Science, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Janice Kim
- Department of Pharmaceutical Science, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Darrell Sawmiller
- MegaNano BioTech, Inc., 3802 Spectrum Blvd. Suite 122, Tampa, FL 33612, USA
| | - Chuanhai Cao
- Department of Pharmaceutical Science, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33612, USA
- USF-Health Byrd Alzheimer’s Institute, University of South Florida, Tampa, FL 33613, USA
- Correspondence:
| |
Collapse
|
8
|
Interactions between apolipoprotein E, sex, and amyloid-beta on cerebrospinal fluid p-tau levels in the European prevention of Alzheimer's dementia longitudinal cohort study (EPAD LCS). EBioMedicine 2022; 83:104241. [PMID: 36041266 PMCID: PMC9440380 DOI: 10.1016/j.ebiom.2022.104241] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 08/11/2022] [Accepted: 08/11/2022] [Indexed: 11/22/2022] Open
Abstract
Background Alzheimer's Disease, the leading cause of dementia, is over-represented in females. The apolipoprotein E (APOE)ε4 allele is the strongest genetic risk factor for late-onset AD and is associated with aberrant cerebrospinal fluid levels (CSF) of total tau (t-tau), phosphorylated tau (p-tau), and amyloid-β (Aβ). There is some evidence that sex may mediate the relationship between APOE status and CSF tau, however, evidence is mixed. Methods We aimed to examine the interaction between sex, APOE ε4 status, CSF Aβ on t-tau and p-tau in 1599 mid-to-late life individuals without a diagnosis of dementia in the European Prevention of Alzheimer's Dementia (EPAD) longitudinal cohort study. Findings We found a significant interaction between APOE status, sex, and CSF Aβ on CSF p-tau levels (β = 0·18, p = 0·04). Specifically, there was a stronger association between APOE status and CSF Aβ42 on CSF p-tau in males compared to females. Further, in females with high Aβ levels (reflecting less cortical deposition), ε4 carriers had significantly elevated p-tau levels relative to non-carriers (W = 39663, p = 0·01). However, there were no significant differences in p-tau between male ε4 carriers and non-carriers with high Aβ (W = 23523, p = 0·64). Interpretation An interaction between sex and cerebrospinal fluid Aβ may mediate the relationship between APOE status and CSF p-tau. These data suggest tau accumulation may be independent of Aβ in females, but not males. Funding Innovative Medicines Initiative, Swedish Research Council, Alzheimer Drug Discovery Foundation, Swedish Alzheimer Foundation, the Swedish state under the agreement between the Swedish government and the County Councils: the ALF-agreement, and the Alzheimer's Association 2021 Zenith Award.
Collapse
|
9
|
Pagni G, Tagliarini C, Carbone MG, Imbimbo BP, Marazziti D, Pomara N. Different Sides of Depression in the Elderly: An In-depth View on the Role of Aβ Peptides. Curr Med Chem 2021; 29:5731-5757. [PMID: 34547994 DOI: 10.2174/0929867328666210921164816] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 08/08/2021] [Accepted: 08/18/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Late-onset depression (LOD) is the most common neuropsychiatric disorder associated with Alzheimer's disease (AD), often associated with structural and functional brain changes, neuropsychological impairments and negative family history for affective disorders. LOD could be a risk factor or a prodromal phase of AD; this has led to the investigation of the link between depression and amyloid-β (Aβ) peptides by measuring Aβ levels in plasma, cerebrospinal fluid (CSF) and brains of elderly depressed subjects. OBJECTIVE Clarify the complex relationship between depression, Aβ peptides and AD. METHOD We evaluated all articles published up to 2019 in PubMed in which Aβ was measured in serum (or plasma), CSF or brain in elderly with Major Depressive Disorder or depressive symptoms evaluated with standard scales. RESULTS Low plasma Aβ42 levels are strongly associated with depression severity. Plasma Aβ40 levels are higher in younger depressed, drug-resistant and those with more severe symptoms. CSF Aβ42 levels are lower in depressed than controls. PET-detected global and region-specific increases in Aβ deposition are sometimes associated with LOD, cognitive impairment, anxiety but not with Cardiovascular Diseases (CVDs)/CVD risk factors. Elderly depressed with CVDs/CVD risk factors have more frequently high plasma Aβ40 levels and drug-resistance; those without these co-morbidities have low plasma Aβ42 levels and a greater cognitive impairment. CONCLUSION Two specific Aβ profiles emerge in elderly depressed. One is associated with Aβ42 reductions in plasma and CSF, possibly reflecting increased brain amyloid deposition and prodromal AD. The other one is characterized by high plasma Aβ40 levels, cerebrovascular disease and clinically associated with increased AD risk.
Collapse
Affiliation(s)
- Giovann Pagni
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 57, 56100. Italy
| | - Claudia Tagliarini
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 57, 56100. Italy
| | - Manuel Glauco Carbone
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 57, 56100. Italy
| | | | - Donatella Marazziti
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 57, 56100. Italy
| | - Nunzio Pomara
- Geriatric Psychiatry Department, Nathan S. Kline Institute, 140 Old Orangeburg Road, Orangeburg, NY 10962. United States
| |
Collapse
|
10
|
Kang SS, Ahn EH, Liu X, Bryson M, Miller GW, Weinshenker D, Ye K. ApoE4 inhibition of VMAT2 in the locus coeruleus exacerbates Tau pathology in Alzheimer's disease. Acta Neuropathol 2021; 142:139-158. [PMID: 33895869 DOI: 10.1007/s00401-021-02315-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 04/08/2021] [Accepted: 04/17/2021] [Indexed: 01/20/2023]
Abstract
ApoE4 enhances Tau neurotoxicity and promotes the early onset of AD. Pretangle Tau in the noradrenergic locus coeruleus (LC) is the earliest detectable AD-like pathology in the human brain. However, a direct relationship between ApoE4 and Tau in the LC has not been identified. Here we show that ApoE4 selectively binds to the vesicular monoamine transporter 2 (VMAT2) and inhibits neurotransmitter uptake. The exclusion of norepinephrine (NE) from synaptic vesicles leads to its oxidation into the toxic metabolite 3,4-dihydroxyphenyl glycolaldehyde (DOPEGAL), which subsequently activates cleavage of Tau at N368 by asparagine endopeptidase (AEP) and triggers LC neurodegeneration. Our data reveal that ApoE4 boosts Tau neurotoxicity via VMAT2 inhibition, reduces hippocampal volume, and induces cognitive dysfunction in an AEP- and Tau N368-dependent manner, while conversely ApoE3 binds Tau and protects it from cleavage. Thus, ApoE4 exacerbates Tau neurotoxicity by increasing VMAT2 vesicle leakage and facilitating AEP-mediated Tau proteolytic cleavage in the LC via DOPEGAL.
Collapse
Affiliation(s)
- Seong Su Kang
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, 615 Michael St. Whitehead BLDG Room #141, Atlanta, GA, 30322, USA
| | - Eun Hee Ahn
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, 615 Michael St. Whitehead BLDG Room #141, Atlanta, GA, 30322, USA
| | - Xia Liu
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, 615 Michael St. Whitehead BLDG Room #141, Atlanta, GA, 30322, USA
| | - Matthew Bryson
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, 615 Michael St. Whitehead BLDG Room #141, Atlanta, GA, 30322, USA
| | - Gary W Miller
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, USA
| | - David Weinshenker
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Keqiang Ye
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, 615 Michael St. Whitehead BLDG Room #141, Atlanta, GA, 30322, USA.
| |
Collapse
|
11
|
Raj A. Graph Models of Pathology Spread in Alzheimer's Disease: An Alternative to Conventional Graph Theoretic Analysis. Brain Connect 2021; 11:799-814. [PMID: 33858198 DOI: 10.1089/brain.2020.0905] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Background: Graph theory and connectomics are new techniques for uncovering disease-induced changes in the brain's structural network. Most prior studied have focused on network statistics as biomarkers of disease. However, an emerging body of work involves exploring how the network serves as a conduit for the propagation of disease factors in the brain and has successfully mapped the functional and pathological consequences of disease propagation. In Alzheimer's disease (AD), progressive deposition of misfolded proteins amyloid and tau is well-known to follow fiber projections, under a "prion-like" trans-neuronal transmission mechanism, through which misfolded proteins cascade along neuronal pathways, giving rise to network spread. Methods: In this review, we survey the state of the art in mathematical modeling of connectome-mediated pathology spread in AD. Then we address several open questions that are amenable to mathematically precise parsimonious modeling of pathophysiological processes, extrapolated to the whole brain. We specifically identify current formal models of how misfolded proteins are produced, aggregate, and disseminate in brain circuits, and attempt to understand how this process leads to stereotyped progression in Alzheimer's and other related diseases. Conclusion: This review serves to unify current efforts in modeling of AD progression that together have the potential to explain observed phenomena and serve as a test-bed for future hypothesis generation and testing in silico. Impact statement Graph theory is a powerful new approach that is transforming the study of brain processes. There do not exist many focused reviews of the subfield of graph modeling of how Alzheimer's and other dementias propagate within the brain network, and how these processes can be mapped mathematically. By providing timely and topical review of this subfield, we fill a critical gap in the community and present a unified view that can serve as an in silico test-bed for future hypothesis generation and testing. We also point to several open and unaddressed questions and controversies that future practitioners can tackle.
Collapse
Affiliation(s)
- Ashish Raj
- Department of Radiology and Biomedical Imaging, University of California at San Francisco, San Francisco, California, USA
| |
Collapse
|
12
|
Cardoso R, Lemos C, Oliveiros B, Almeida MR, Baldeiras I, Pereira CF, Santos A, Duro D, Vieira D, Santana I, Oliveira CR. APOEɛ4-TOMM40L Haplotype Increases the Risk of Mild Cognitive Impairment Conversion to Alzheimer's Disease. J Alzheimers Dis 2020; 78:587-601. [PMID: 33016906 DOI: 10.3233/jad-200556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Mild cognitive impairment (MCI) has been considered as a pre-dementia stage, although the factors leading to Alzheimer's disease (AD) conversion remain controversial. OBJECTIVE Evaluate whether TOMM40 poly-T (TOMM40' 523) polymorphism is associated with the risk and conversion time from MCI to AD and secondly with AD cerebrospinal fluid (CSF) biomarkers, disentangling the APOE genotype. METHODS 147 AD patients, 102 MCI patients, and 105 cognitively normal controls were genotyped for poly-T polymorphism. MCI patients were subdivided into two groups, the group of patients that converted to AD (MCI-AD) and the group of those that remained stable (MCI-S). RESULTS TOMM40' 523 L allele was significantly more frequent in the MCI-AD group and having at least one L allele significantly increased the risk of conversion from MCI to AD (OR = 8.346, p < 0.001, 95% CI: 2.830 to 24.617). However, when adjusted for the presence of APOEɛ4 allele, both the L allele and ɛ4 allele lost significance in the model (p > 0.05). We then analyzed the APOEɛ4-TOMM40' 523 L haplotype and observed that patients carrying this haplotype had significantly higher risk (OR = 5.83; 95% CI = 2.30-14.83) and mean lower times of conversion to AD (p = 0.003). This haplotype was also significantly associated with a biomarker profile compatible with AD (p = 0.007). CONCLUSION This study shows that the APOEɛ4-TOMM40' 523 L haplotype is associated with a higher risk and shorter times of conversion from MCI to AD, possibly driven by CSF biomarkers and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Remy Cardoso
- Center for Neuroscience and Cell Biology, CNC-CIBB, University of Coimbra, Coimbra, Portugal
| | - Carolina Lemos
- UnIGENe, IBMC -Institute for Molecular and Cell Biology, Porto, Portugal.,i3S -Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,ICBAS - Instituto Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Bárbara Oliveiros
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Laboratory of Biostatistics and Medical Informatics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Maria Rosário Almeida
- Center for Neuroscience and Cell Biology, CNC-CIBB, University of Coimbra, Coimbra, Portugal
| | - Inês Baldeiras
- Center for Neuroscience and Cell Biology, CNC-CIBB, University of Coimbra, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Neurochemistry Laboratory, Neurology Department, Coimbra University Hospital (CHUC), Coimbra, Portugal
| | - Cláudia Fragão Pereira
- Center for Neuroscience and Cell Biology, CNC-CIBB, University of Coimbra, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Ana Santos
- Center for Neuroscience and Cell Biology, CNC-CIBB, University of Coimbra, Coimbra, Portugal
| | - Diana Duro
- Neurology Department, Coimbra University Hospital (CHUC), Coimbra, Portugal
| | - Daniela Vieira
- Neurology Department, Coimbra University Hospital (CHUC), Coimbra, Portugal
| | - Isabel Santana
- Center for Neuroscience and Cell Biology, CNC-CIBB, University of Coimbra, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Neurology Department, Coimbra University Hospital (CHUC), Coimbra, Portugal
| | - Catarina Resende Oliveira
- Center for Neuroscience and Cell Biology, CNC-CIBB, University of Coimbra, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Clinical Academic Center of Coimbra, Coimbra, Portugal
| |
Collapse
|
13
|
Babapour Mofrad R, Tijms BM, Scheltens P, Barkhof F, van der Flier WM, Sikkes SA, Teunissen CE. Sex differences in CSF biomarkers vary by Alzheimer disease stage and APOE ε4 genotype. Neurology 2020; 95:e2378-e2388. [DOI: 10.1212/wnl.0000000000010629] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 05/19/2020] [Indexed: 12/25/2022] Open
Abstract
ObjectiveTo evaluate sex differences in CSF biomarkers, taking the potential modifying role of clinical disease stage and APOE ε4 genotype into account.MethodWe included participants (n = 1,801) with probable Alzheimer disease (AD) dementia (n = 937), mild cognitive impairment (MCI; n = 437), and subjective cognitive decline (SCD; n = 427). Main outcomes were CSF β-amyloid1–42 (Aβ42), total tau (t-Tau), and tau phosphorylated at threonine 181 (p-Tau) levels. Age-corrected 3-way interactions between sex, disease stage (i.e., syndrome diagnosis at baseline), and APOE ε4 were tested with linear regression analyses for each outcome measure. In case of significant interactions (p < 0.05), sex differences were further evaluated by stratifying analyses for clinical disease stage and APOE ε4 genotype, including age as a covariate.ResultsThree-way interactions were significant for t-Tau (p < 0.001) and p-Tau (p < 0.01) but not Aβ42. In APOE ε4 carriers, women showed higher p-Tau concentrations than men in SCD (Cohen d [95% confidence interval]: t-Tau = 0.52 [0.19–0.84], p < 0.001; p-Tau = 0.44 [0.11–0.77] p = 0.004) and MCI (Cohen d [95% CI]: t-Tau = 0.54 [0.28–0.80], p < 0.001; p-Tau = 0.52 [0.26–0.77], p < 0.001) but not in AD dementia. In APOE ε4 noncarriers, women showed higher p-Tau concentrations in MCI (Cohen d [95% CI]: t-Tau = 0.49 [0.17–0.80], p = 0.002; p-Tau = 0.47 [0.16–0.78], p = 0.003) and AD dementia (Cohen d [95% CI]: t-Tau = 0.42 [0.19–0.65], p < 0.001; p-Tau = 0.38 [0.15–0.61] p = 0.002) but not in SCD.ConclusionsWithin APOE ε4 carriers, sex differences in CSF p-Tau are more evident in early disease stages, whereas for APOE ε4 noncarriers, sex differences are more evident in advanced disease stages. These findings suggest that the effect of APOE ε4 on sex differences in CSF biomarkers depends on disease stage in AD.
Collapse
|
14
|
Relationship between Urinary Alzheimer-Associated Neuronal Thread Protein and Apolipoprotein Epsilon 4 Allele in the Cognitively Normal Population. Neural Plast 2020; 2020:9742138. [PMID: 32587611 PMCID: PMC7294364 DOI: 10.1155/2020/9742138] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 04/18/2020] [Accepted: 05/22/2020] [Indexed: 12/23/2022] Open
Abstract
We investigated the relationship between urinary Alzheimer-associated neuronal thread protein (AD7c-NTP) levels and apolipoprotein epsilon 4 (ApoE ɛ4) alleles, as well as other factors that cause cognitive decline, in the cognitively normal population. We recruited 329 cognitively normal right-handed Han Chinese subjects who completed ApoE gene testing and urinary AD7c-NTP testing. There was no significant difference in urinary AD7c-NTP levels between the normal control and subjective cognitive decline groups. Urinary AD7c-NTP levels were significantly higher in subjects with ApoE ɛ3/4 and 4/4 [0.6074 (0.6541) ng/mL] than in subjects without ApoE ɛ4 [0.4368 (0.3392) ng/mL and 0.5287 (0.3656) ng/mL], and urinary AD7c-NTP levels positively correlated with ApoE genotype grade (r = 0.165, p = 0.003). There were significant differences in urinary AD7c-NTP levels between subjects with and without a history of coronary heart disease or diabetes. Urinary AD7c-NTP levels were not related to years of education, nature of work, family history of dementia, a history of hypertension, stroke, anemia, or thyroid dysfunction. Urinary AD7c-NTP levels were positively correlated with ApoE grade in the cognitively normal population. The relationship between risk factors of cognitive decline and urinary AD7c-NTP levels provides a new way for us to understand AD and urinary AD7c-NTP.
Collapse
|
15
|
López-Cuenca I, de Hoz R, Salobrar-García E, Elvira-Hurtado L, Rojas P, Fernández-Albarral JA, Barabash A, Salazar JJ, Ramírez AI, Ramírez JM. Macular Thickness Decrease in Asymptomatic Subjects at High Genetic Risk of Developing Alzheimer's Disease: An OCT Study. J Clin Med 2020; 9:jcm9061728. [PMID: 32503282 PMCID: PMC7355697 DOI: 10.3390/jcm9061728] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 05/29/2020] [Accepted: 06/01/2020] [Indexed: 12/17/2022] Open
Abstract
In this case control study, we examined the retinal thickness of the different layers in the macular region and peripapillary retinal nerve fiber layer (RNFL) with optical coherence tomography (OCT) in healthy cognitive subjects (from 51 to 74 years old) at high genetic risk for developing Alzheimer’s disease (AD). Thirty-five subjects with a family history of Alzheimer disease (AD) (FH+) and ApoE ɛ4 carriers and 29 age-matched control subjects without a family history of AD (FH−) and ApoE ɛ4 non-carriers were included. Compared to FH− ApoE ɛ4 non-carriers, in FH+ ApoE ɛ4 carriers, there were statistically significant decreases (p < 0.05) in (i) the foveal area of mRNFL; (ii) the inferior and nasal sectors in the outer and inner macular ring in the inner plexiform layer (IPL); (iii) the foveal area and the inferior sector in the outer macular ring in the inner nuclear layer (INL); and (iv) the inferior sector of the outer macular ring in the outer plexiform layer (OPL). However, no statistically significant differences were found in the peripapillary thickness of RNFL between both study groups. In subjects with cognitive health and high genetic risk for the development of AD, initial changes appeared in the macular area. OCT could be a promising, cost-effective and non-invasive test useful in early AD, before the onset of clinical symptoms.
Collapse
Affiliation(s)
- Inés López-Cuenca
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Universidad Complutense de Madrid (UCM), 28040 Madrid, Spain; (I.L.-C.); (R.d.H.); (E.S.-G.); (L.E.-H.); (P.R.); (J.A.F.-A.); (J.J.S.)
| | - Rosa de Hoz
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Universidad Complutense de Madrid (UCM), 28040 Madrid, Spain; (I.L.-C.); (R.d.H.); (E.S.-G.); (L.E.-H.); (P.R.); (J.A.F.-A.); (J.J.S.)
- IIORC, Faculty of Medicine, 28011 Madrid, Spain
- Facultad de Óptica y Optometría, Departamento de Inmunología, Oftalmología y ORL, UCM, 28037 Madrid, Spain
| | - Elena Salobrar-García
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Universidad Complutense de Madrid (UCM), 28040 Madrid, Spain; (I.L.-C.); (R.d.H.); (E.S.-G.); (L.E.-H.); (P.R.); (J.A.F.-A.); (J.J.S.)
- IIORC, Faculty of Medicine, 28011 Madrid, Spain
- Facultad de Óptica y Optometría, Departamento de Inmunología, Oftalmología y ORL, UCM, 28037 Madrid, Spain
| | - Lorena Elvira-Hurtado
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Universidad Complutense de Madrid (UCM), 28040 Madrid, Spain; (I.L.-C.); (R.d.H.); (E.S.-G.); (L.E.-H.); (P.R.); (J.A.F.-A.); (J.J.S.)
| | - Pilar Rojas
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Universidad Complutense de Madrid (UCM), 28040 Madrid, Spain; (I.L.-C.); (R.d.H.); (E.S.-G.); (L.E.-H.); (P.R.); (J.A.F.-A.); (J.J.S.)
- Hospital General Universitario Gregorio Marañón, Instituto Oftálmico de Madrid, 28007 Madrid, Spain
| | - José A. Fernández-Albarral
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Universidad Complutense de Madrid (UCM), 28040 Madrid, Spain; (I.L.-C.); (R.d.H.); (E.S.-G.); (L.E.-H.); (P.R.); (J.A.F.-A.); (J.J.S.)
| | - Ana Barabash
- Endocrinology and Nutrition Department, Hospital Clínico Universitario San Carlos and Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, 28040 Madrid, Spain;
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas, 28029 Madrid, Spain
| | - Juan J. Salazar
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Universidad Complutense de Madrid (UCM), 28040 Madrid, Spain; (I.L.-C.); (R.d.H.); (E.S.-G.); (L.E.-H.); (P.R.); (J.A.F.-A.); (J.J.S.)
- IIORC, Faculty of Medicine, 28011 Madrid, Spain
- Facultad de Óptica y Optometría, Departamento de Inmunología, Oftalmología y ORL, UCM, 28037 Madrid, Spain
| | - Ana I. Ramírez
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Universidad Complutense de Madrid (UCM), 28040 Madrid, Spain; (I.L.-C.); (R.d.H.); (E.S.-G.); (L.E.-H.); (P.R.); (J.A.F.-A.); (J.J.S.)
- IIORC, Faculty of Medicine, 28011 Madrid, Spain
- Facultad de Óptica y Optometría, Departamento de Inmunología, Oftalmología y ORL, UCM, 28037 Madrid, Spain
- Correspondence: (A.I.R.); (J.M.R.)
| | - José M. Ramírez
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Universidad Complutense de Madrid (UCM), 28040 Madrid, Spain; (I.L.-C.); (R.d.H.); (E.S.-G.); (L.E.-H.); (P.R.); (J.A.F.-A.); (J.J.S.)
- IIORC, Faculty of Medicine, 28011 Madrid, Spain
- Facultad de Medicina, Departamento de Inmunología, Oftalmología y ORL, UCM, 28040 Madrid, Spain
- Correspondence: (A.I.R.); (J.M.R.)
| |
Collapse
|
16
|
Zaldua S, Damen FC, Pisharody R, Thomas R, Fan KD, Ekkurthi GK, Scheinman SB, Alahmadi S, Marottoli FM, Alford S, Cai K, Tai LM. Epidermal growth factor treatment of female mice that express APOE4 at an age of advanced pathology mitigates behavioral and cerebrovascular dysfunction. Heliyon 2020; 6:e03919. [PMID: 32478184 PMCID: PMC7251379 DOI: 10.1016/j.heliyon.2020.e03919] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 11/18/2019] [Accepted: 04/30/2020] [Indexed: 02/01/2023] Open
Abstract
APOE4 is a major genetic risk factor for Alzheimer's disease and high amyloid-β (Aβ) levels in the brain are a pathological hallmark of the disease. However, the contribution of specific APOE-modulated Aβ-dependent and Aβ-independent functions to cognitive decline remain unclear. Increasing evidence supports a role of APOE in modulating cerebrovascular function, however whether ameliorating this dysfunction can improve behavioral function is still under debate. We have previously demonstrated that systemic epidermal growth factor (EGF) treatment, which is important for vascular function, at early stages of pathology (treatment from 6 to 8 months) is beneficial for recognition and spatial memory and cerebrovascular function in female mice that express APOE4. These data raise the important question of whether EGF can improve APOE4-associated cerebrovascular and behavioral dysfunction when treatment is initiated at an age of advanced pathology. Positive findings would support the development of therapies that target cerebrovascular dysfunction associated with APOE4 in aging and AD in individuals with advanced cognitive impairment. Therefore, in this study female mice that express APOE4 in the absence (E4FAD- mice) or presence (E4FAD+ mice) of Aβ overproduction were treated from 8 to 10 months of age systemically with EGF. EGF treatment mitigated behavioral dysfunction in recognition memory and spatial learning and improved hippocampal neuronal function in both E4FAD+ and E4FAD- mice, suggesting that EGF treatment improves Aβ-independent APOE4-associated deficits. The beneficial effects of EGF treatment on behavior occurred in tandem with improved markers of cerebrovascular function, including lower levels of fibrinogen, lower permeability when assessed by MRI and higher percent area coverage of laminin and CD31 in the hippocampus. These data suggest a mechanistic link among EGF signaling, cerebrovascular function and APOE4-associated behavioral deficits in mice with advanced AD-relevant pathology.
Collapse
Affiliation(s)
- Steve Zaldua
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Frederick C Damen
- Department of Radiology, University of Illinois Hospital & Health Sciences System, Suite 103, 2242 West Harrison Street, Chicago, IL 60612, USA
| | - Rohan Pisharody
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Riya Thomas
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Kelly D Fan
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Giri K Ekkurthi
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Sarah B Scheinman
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Sami Alahmadi
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Felecia M Marottoli
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Simon Alford
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Kejia Cai
- Department of Radiology, University of Illinois Hospital & Health Sciences System, Suite 103, 2242 West Harrison Street, Chicago, IL 60612, USA
- Department of Bioengineering, University of Illinois Hospital & Health Sciences System, Suite 103, 2242 West Harrison Street, Chicago, IL 60612, USA
| | - Leon M Tai
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| |
Collapse
|
17
|
Li XL, Zhan RQ, Zheng W, Jiang H, Zhang DF, Shen XL. Positive association between soil arsenic concentration and mortality from alzheimer's disease in mainland China. J Trace Elem Med Biol 2020; 59:126452. [PMID: 31962196 PMCID: PMC7350902 DOI: 10.1016/j.jtemb.2020.126452] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 12/11/2019] [Accepted: 01/08/2020] [Indexed: 12/22/2022]
Abstract
OBJECTIVES The current study was designed to investigate the relationship between the soil arsenic (As) concentration and the mortality from Alzheimer's disease (AD) in mainland China. STUDY DESIGN Ecological study. METHODS Twenty-two provinces and 3 municipal districts in mainland China were included in this study. The As concentrations in soil in 1990 was obtained from the China State Environmental Protection Bureau; the data on annual mortality of AD from 1991 to 2000 were obtained from the National Death Cause Surveillance Database of China. Using these data, we calculated the spearman correlation coefficient between soil As concentration and AD mortality, and the relative risk (RR) between soil As levels and AD mortality by quartile-dividing study groups. RESULTS The spearman correlation coefficient between As concentration and AD mortality was 0.552 (p = 0.004), 0.616 (p = 0.001) and 0.622 (p = 0.001) in the A soil As (eluvial horizon), the C soil As (parent material horizon), and the Total soil As (A soil As + C soil As), respectively. When the A soil As concentration was over 9.05 mg/kg, 10.40 mg/kg and 13.10 mg/kg, the relative risk was 0.835 (95 % CI: 0.832, 0.838), 1.969 (95 %CI: 1.955, 1.982), and 2.939 (95 % CI: 2.920, 2.958), respectively; when the C soil As reached 9.45 mg/kg, 11.10 mg/kg and 13.55 mg/kg, the relative risk was 4.349 (95 % CI: 4.303, 4.396), 6.108 (95 % CI: 6.044, 6.172), and 9.125 (95 %CI: 9.033, 9.219), respectively. No correlation was found between lead, cadmium, and mercury concentration in the soil and AD mortality. CONCLUSION There was an apparent soil As concentration dependent increase in AD mortality. Results of this study may provide evidence for a possible causal linkage between arsenic exposure and the death risk from AD.
Collapse
Affiliation(s)
- Xue-Lian Li
- Department of Epidemiology and Health Statistics, Medical College of Qingdao University, Qingdao, China
| | - Run-Qing Zhan
- Qingdao University Affiliated Hiser Hospital, Qingdao, China
| | - Wei Zheng
- School of Health Sciences, Purdue University, West Lafayette, IN, USA
| | - Hong Jiang
- Department of Physiology, Medical College of Qingdao University, Qingdao, China
| | - Dong-Feng Zhang
- Department of Epidemiology and Health Statistics, Medical College of Qingdao University, Qingdao, China
| | - Xiao-Li Shen
- Department of Epidemiology and Health Statistics, Medical College of Qingdao University, Qingdao, China.
| |
Collapse
|
18
|
Zhou R, Chen H, Ye F, Huang S, Zhang J. Influence of Hypertension on Longitudinal Changes in Brain Glucose Metabolism Was Modified by the APOE4 Allele Among Cognitively Normal Older Individuals. Front Aging Neurosci 2020; 12:85. [PMID: 32308617 PMCID: PMC7146026 DOI: 10.3389/fnagi.2020.00085] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 03/12/2020] [Indexed: 02/05/2023] Open
Abstract
Objective To examine whether the influence of hypertension (HTN) status on longitudinal changes in brain glucose metabolism was modified by the apolipoprotein 4 (APOE4) status among older people with normal cognition. Methods In this study, we included 217 older individuals with normal cognition from the Alzheimer’s Disease Neuroimaging Initiative (ADNI) study. Participants were divided into the HTN and no HTN groups based on self-reported medical history. Brain glucose metabolism was assessed by 18F-fluorodeoxyglucose-positron emission tomography (FDG-PET). Linear mixed model was fitted to examine the association between the HTN × APOE4 interaction and longitudinal changes in brain glucose metabolism after controlling for several covariates. Results In the present study, we found that the association between HTN status and longitudinal changes in brain glucose metabolism varied as a function of the APOE4 status, such that the HTN/APOE4+ group showed a steeper decline in FDG SUVR than all other groups (No HTN/APOE4-, HTN/APOE4-, and No HTN/APOE4+). Nevertheless, there was no significant difference in the rate of decline in FDG SUVR among other groups (No HTN/APOE4-, HTN/APOE4-, and No HTN/APOE4+). Conclusion The APOE4 genotype interacted with hypertension status to affect longitudinal changes in brain glucose metabolism among older individual with normal cognition, such that the HTN/APOE4+ group showed a steeper decline in FDG SUVR than other groups.
Collapse
Affiliation(s)
- Rui Zhou
- Department of Cardiology, Wenzhou People's Hospital, Wenzhou, China
| | - Hao Chen
- Department of Cardiology, Wenzhou People's Hospital, Wenzhou, China
| | - Fanhao Ye
- Department of Cardiology, Wenzhou People's Hospital, Wenzhou, China
| | - Shiwei Huang
- Department of Cardiology, Wenzhou People's Hospital, Wenzhou, China
| | - Jie Zhang
- Independent Researcher, Hangzhou, China
| |
Collapse
|
19
|
Synergistic effects of APOE and sex on the gut microbiome of young EFAD transgenic mice. Mol Neurodegener 2019; 14:47. [PMID: 31861986 PMCID: PMC6923910 DOI: 10.1186/s13024-019-0352-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 12/11/2019] [Indexed: 12/14/2022] Open
Abstract
Background Alzheimer’s disease (AD) is a fatal neurodegenerative disease. APOE4 is the greatest genetic risk factor for AD, increasing risk up to 15-fold compared to the common APOE3. Importantly, female (♀) APOE4 carriers have a greater risk for developing AD and an increased rate of cognitive decline compared to male (♂) APOE4 carriers. While recent evidence demonstrates that AD, APOE genotype, and sex affect the gut microbiome (GM), how APOE genotype and sex interact to affect the GM in AD remains unknown. Methods This study analyzes the GM of 4-month (4 M) ♂ and ♀ E3FAD and E4FAD mice, transgenic mice that overproduce amyloid-β 42 (Aβ42) and express human APOE3+/+ or APOE4+/+. Fecal microbiotas were analyzed using high-throughput sequencing of 16S ribosomal RNA gene amplicons and clustered into operational taxonomic units (OTU). Microbial diversity of the EFAD GM was compared across APOE, sex and stratified by APOE + sex, resulting in 4-cohorts (♂E3FAD, ♀E3FAD, ♂E4FAD and ♀E4FAD). Permutational multivariate analysis of variance (PERMANOVA) evaluated differences in bacterial communities between cohorts and the effects of APOE + sex. Mann-Whitney tests and machine-learning algorithms identified differentially abundant taxa associated with APOE + sex. Results Significant differences in the EFAD GM were associated with APOE genotype and sex. Stratification by APOE + sex revealed that APOE-associated differences were exhibited in ♂EFAD and ♀EFAD mice, and sex-associated differences were exhibited in E3FAD and E4FAD mice. Specifically, the relative abundance of bacteria from the genera Prevotella and Ruminococcus was significantly higher in ♀E4FAD compared to ♀E3FAD, while the relative abundance of Sutterella was significantly higher in ♂E4FAD compared to ♂E3FAD. Based on 29 OTUs identified by the machine-learning algorithms, heatmap analysis revealed significant clustering of ♀E4FAD separate from other cohorts. Conclusions The results demonstrate that the 4 M EFAD GM is modulated by APOE + sex. Importantly, the effect of APOE4 on the EFAD GM is modulated by sex, a pattern similar to the greater AD pathology associated with ♀E4FAD. While this study demonstrates the importance of interactive effects of APOE + sex on the GM in young AD transgenic mice, changes associated with the development of pathology remain to be defined.
Collapse
|
20
|
Abstract
Currently established and employed biomarkers of Alzheimer's disease (AD) predominantly mirror AD-associated molecular and structural brain changes. While they are necessary for identifying disease-specific neuropathology, they lack a clear and robust relationship with the clinical presentation of dementia; they can be altered in healthy individuals, while they often inadequately mirror the degree of cognitive and functional deficits in affected subjects. There is growing evidence that synaptic loss and dysfunction are early events during the trajectory of AD pathogenesis that best correlate with the clinical symptoms, suggesting measures of brain functional deficits as candidate early markers of AD. Resting-state electroencephalography (EEG) is a widely available and noninvasive diagnostic method that provides direct insight into brain synaptic activity in real time. Quantitative EEG (qEEG) analysis additionally provides information on physiologically meaningful frequency components, dynamic alterations and topography of EEG signal generators, i.e. neuronal signaling. Numerous studies have shown that qEEG measures can detect disruptions in activity, topographical distribution and synchronization of neuronal (synaptic) activity such as generalized EEG slowing, reduced global synchronization and anteriorization of neuronal generators of fast-frequency resting-state EEG activity in patients along the AD continuum. Moreover, qEEG measures appear to correlate well with surrogate markers of AD neuropathology and discriminate between different types of dementia, making them promising low-cost and noninvasive markers of AD. Future large-scale longitudinal clinical studies are needed to elucidate the diagnostic and prognostic potential of qEEG measures as early functional markers of AD on an individual subject level.
Collapse
Affiliation(s)
- Una Smailovic
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden.
| | - Vesna Jelic
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Clinic for Cognitive Disorders, Theme Aging, Karolinska University Hospital, Huddinge, Sweden
| |
Collapse
|
21
|
Damen FC, Tain RW, Thomas R, Li W, Tai L, Cai K. Evaluation of B 0-correction of relative CBF maps using tagging distance dependent Z-spectrum (TADDZ). Magn Reson Imaging 2019; 65:83-89. [PMID: 31669538 DOI: 10.1016/j.mri.2019.10.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 08/27/2019] [Accepted: 10/08/2019] [Indexed: 12/17/2022]
Abstract
Arterial spin labeling (ASL) MRI, based on endogenous contrast from blood water, is used in research and diagnosis of cerebral vascular conditions. However, artifacts due to imperfect imaging conditions such as B0-inhomogeneity (ΔB0) could lead to variations in the quantification of relative cerebral blood flow (CBF). In this study, we evaluate a new approach using tagging distance dependent Z-spectrum (TADDZ) data, similar to the ΔB0 corrections in the chemical exchange saturation transfer (CEST) experiments, to remove the imaging plane B0 inhomogeneity induced CBF artifacts in ASL MRI. Our results indicate that imaging-plane B0-inhomogeneity can lead to variations and errors in the relative CBF maps especially under small tagging distances. Along with an acquired B0 map, TADDZ data helps to eliminate B0-inhomogeneity induced artifacts in the resulting relative CBF maps. We demonstrated the effective use of TADDZ data to reduce variation while subjected to systematic changes in ΔB0. In addition, TADDZ corrected ASL MRI, with improved consistency, was shown to outperform conventional ASL MRI by differentiating the subtle CBF difference in Alzheimer's disease (AD) mice brains with different APOE genotypes.
Collapse
Affiliation(s)
- Frederick C Damen
- Department of Radiology, University of Illinois at Chicago, Chicago, IL, United States
| | - Rong-Wen Tain
- Department of Radiology, University of Illinois at Chicago, Chicago, IL, United States; Brain Imaging Research, University of California, Irvine, CA, United States
| | - Riya Thomas
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Weigo Li
- Research Resources Center, University of Illinois at Chicago, Chicago, IL, United States; Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, United States; Department of Radiology, Northwestern University, Chicago, IL, United States
| | - Leon Tai
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Kejia Cai
- Department of Radiology, University of Illinois at Chicago, Chicago, IL, United States; Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, United States; Center for MR Research, University of Illinois at Chicago, Chicago, IL, United States.
| |
Collapse
|
22
|
Yuan T, Chu C, Shi R, Cui T, Zhang X, Zhao Y, Shi X, Hui Y, Pan J, Qian R, Dai X, Liu Z, Liu X. ApoE-Dependent Protective Effects of Sesamol on High-Fat Diet-Induced Behavioral Disorders: Regulation of the Microbiome-Gut-Brain Axis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:6190-6201. [PMID: 31117496 DOI: 10.1021/acs.jafc.9b01436] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Sesamol, an antioxidant lignan from sesame oil, possesses neuroprotective bioactivities. The present work was aimed to elucidate the systemic protective effects of sesamol on cognitive deficits and to determine the possible link between gut and brain. Wildtype and ApoE-/- mice were treated with a high-fat diet and sesamol (0.05%, w/v, in drinking water) for 10 weeks. Behavioral tests including Morris-water maze, Y-maze, and elevated plus maze tests indicated that sesamol could only improve cognitive deficits and anxiety behaviors in wildtype. Consistently, sesamol improved synapse ultrastructure and inhibited Aβ accumulation in an ApoE-dependent manner. Moreover, sesamol prevented dietary-induced gut barrier damages and systemic inflammation. Sesamol also reshaped gut microbiome and improved the generation of microbial metabolites short-chain fatty acids. To summarize, this study revealed that the possible mechanism of neuroprotective effects of sesamol might be ApoE-dependent, and its beneficial effects on gut microbiota/metabolites could be translated into neurodegenerative diseases treatment.
Collapse
Affiliation(s)
- Tian Yuan
- College of Food Science and Engineering , Northwest A&F University , Yangling , China
| | - Chuanqi Chu
- School of Food Science and Technology , Jiangnan University , Wuxi , China
| | - Rubing Shi
- College of Food Science and Engineering , Northwest A&F University , Yangling , China
| | - Tianlin Cui
- College of Food Science and Engineering , Northwest A&F University , Yangling , China
| | - Xinglin Zhang
- College of Food Science and Engineering , Northwest A&F University , Yangling , China
| | - Yihang Zhao
- College of Food Science and Engineering , Northwest A&F University , Yangling , China
| | - Xu Shi
- College of Food Science and Engineering , Northwest A&F University , Yangling , China
| | - Yan Hui
- Department of Food Science , University of Copenhagen , Copenhagen , Denmark
| | - Junru Pan
- College of Food Science and Engineering , Northwest A&F University , Yangling , China
| | - Rui Qian
- Food Analysis and Development Center , Beijing ZhiYunDa Technology, Co., LTD. , Beijing , China
| | - Xiaoshuang Dai
- BGI Institute of Applied Agriculture , BGI-Shenzhen , Shenzhen , China
| | - Zhigang Liu
- College of Food Science and Engineering , Northwest A&F University , Yangling , China
| | - Xuebo Liu
- College of Food Science and Engineering , Northwest A&F University , Yangling , China
| |
Collapse
|
23
|
Kou X, Chen D, Chen N. Physical Activity Alleviates Cognitive Dysfunction of Alzheimer's Disease through Regulating the mTOR Signaling Pathway. Int J Mol Sci 2019; 20:ijms20071591. [PMID: 30934958 PMCID: PMC6479697 DOI: 10.3390/ijms20071591] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 03/27/2019] [Accepted: 03/27/2019] [Indexed: 02/08/2023] Open
Abstract
Alzheimer's disease (AD) is one of the most common aging-related progressive neurodegenerative disorders, and can result in great suffering for a large portion of the aged population. Although the pathogenesis of AD is being elucidated, the exact mechanisms are still unclear, thereby impeding the development of effective drugs, supplements, and other interventional strategies for AD. In recent years, impaired autophagy associated with microRNA (miRNA) dysfunction has been reported to be involved in aging and aging-related neurodegenerative diseases. Therefore, miRNA-mediated regulation for the functional status of autophagy may become one of the potent interventional strategies for AD. Mounting evidence from in vivo AD models has demonstrated that physical activity can exert a neuroprotective role in AD. In addition, autophagy is strictly regulated by the mTOR signaling pathway. In this article, the regulation of the functional status of autophagy through the mTOR signaling pathway during physical activity is systematically discussed for the prevention and treatment of AD. This concept will be beneficial to developing novel and effective targets that can create a direct link between pharmacological intervention and AD in the future.
Collapse
Affiliation(s)
- Xianjuan Kou
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Health Science, Wuhan Sports University, Wuhan 430079, China.
| | - Dandan Chen
- Graduate School, Wuhan Sports University, Wuhan 430079, China.
| | - Ning Chen
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Health Science, Wuhan Sports University, Wuhan 430079, China.
| |
Collapse
|
24
|
Abstract
BACKGROUND The growing body of evidence indicating the heterogeneity of Alzheimer's disease (AD), coupled with disappointing clinical studies directed at a fit-for-all therapy, suggest that the development of a single magic cure suitable for all cases may not be possible. This calls for a shift in paradigm where targeted treatment is developed for specific AD subpopulations that share distinct genetic or pathological properties. Apolipoprotein E4 (apoE4), the most prevalent genetic risk factor of AD, is expressed in more than half of AD patients and is thus an important possible AD therapeutic target. REVIEW This review focuses initially on the pathological effects of apoE4 in AD, as well as on the corresponding cellular and animal models and the suggested cellular and molecular mechanisms which mediate them. The second part of the review focuses on recent apoE4-targeted (from the APOE gene to the apoE protein and its interactors) therapeutic approaches that have been developed in animal models and are ready to be translated to human. Further, the issue of whether the pathological effects of apoE4 are due to loss of protective function or due to gain of toxic function is discussed herein. It is possible that both mechanisms coexist, with certain constituents of the apoE4 molecule and/or its downstream signaling mediating a toxic effect, while others are associated with a loss of protective function. CONCLUSION ApoE4 is a promising AD therapeutic target that remains understudied. Recent studies are now paving the way for effective apoE4-directed AD treatment approaches.
Collapse
|
25
|
Chen R, Lee C, Lin X, Zhao C, Li X. Novel function of VEGF-B as an antioxidant and therapeutic implications. Pharmacol Res 2019; 143:33-39. [PMID: 30851357 DOI: 10.1016/j.phrs.2019.03.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 03/01/2019] [Accepted: 03/01/2019] [Indexed: 12/14/2022]
Abstract
Oxidative stress, due to insufficiency of antioxidants or over-production of oxidants, can lead to severe cell and tissue damage. Oxidative stress occurs constantly and has been shown to be involved in innumerable diseases, such as degenerative, cardiovascular, neurological, and metabolic disorders, cancer, and aging, thus highlighting the vital need of antioxidant defense mechanisms. Vascular endothelial growth factor B (VEGF-B) was discovered a long time ago, and is abundantly expressed in most types of cells and tissues. VEGF-B remained functionally mysterious for many years and later on has been shown to be minimally angiogenic. Recently, VEGF-B is reported to be a potent antioxidant by boosting the expression of key antioxidant enzymes. Thus, one major role of VEGF-B lies in safeguarding tissues and cells from oxidative stress-induced damage. VEGF-B may therefore have promising therapeutic utilities in treating oxidative stress-related diseases. In this review, we discuss the current knowledge on the newly discovered antioxidant function of VEGF-B and the related molecular mechanisms, particularly, in relationship to some oxidative stress-related diseases, such as retinitis pigmentosa, age-related macular degeneration, diabetic retinopathy, glaucoma, amyotrophic lateral sclerosis, Alzheimer's disease, and Parkinson's disease.
Collapse
Affiliation(s)
- Rongyuan Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Chunsik Lee
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Xianchai Lin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Chen Zhao
- Eye Institute, Eye and ENT Hospital, Shanghai Medical College, Fudan University, China; Key Laboratory of Myopia of State Health Ministry (Fudan University) and Shanghai Key Laboratory of Visual Impairment and Restoration, 200023, Shanghai, China.
| | - Xuri Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China.
| |
Collapse
|
26
|
Galle SA, van der Spek A, Drent ML, Brugts MP, Scherder EJA, Janssen JAMJL, Ikram MA, van Duijn CM. Revisiting the Role of Insulin-Like Growth Factor-I Receptor Stimulating Activity and the Apolipoprotein E in Alzheimer's Disease. Front Aging Neurosci 2019; 11:20. [PMID: 30809143 PMCID: PMC6380107 DOI: 10.3389/fnagi.2019.00020] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 01/24/2019] [Indexed: 12/18/2022] Open
Abstract
Background: Alterations in insulin-like growth factor I (IGF-I) signaling have been associated with dementia and Alzheimer's disease (AD). Studies on the association between IGF-I levels and dementia risk have been inconclusive. We reported earlier that higher levels of IGF-I receptor stimulating activity are associated with a higher prevalence and incidence of dementia. Objective: In the present study, we test the robustness of the association between IGF-I receptor stimulating activity and dementia by extending the follow-up period to 16 years and investigate possible effect modification by apolipoprotein E (ApoE). Methods: At baseline, circulating IGF-I receptor stimulating activity was determined by the IGF-I kinase receptor activation (KIRA) assay in 1,014 elderly from the Rotterdam Study. Dementia was assessed from baseline (1997-1999) to follow-up in January 2015. Associations of IGF-I receptor stimulating activity and incident dementia were assessed with Cox proportional hazards models. Results: During 10,752 person-years of follow-up, 174 people developed dementia. In the extended follow-up we no longer observed a dose-response relationship between IGF-I receptor stimulating activity and risk of dementia [adjusted odds ratio 1.11; 95% confidence interval (CI) 0.97-1.28]. Interestingly, we found evidence of an interaction between ApoE-ε4 and tertiles of IGF-I receptor stimulating activity. IGF-I receptor stimulating activity in the median and top tertiles was related to increased dementia incidence in hetero- and homozygotes of the ApoE-ε4 allele, but did not show any association with dementia risk in people without the ApoE-ε4 allele (adjusted odds ratio medium vs. low IGF-I receptor stimulating activity in ApoE-ε4 carriers: 1.45; 95% CI 1.00-2.12). These findings suggest a threshold effect in ApoE-ε4 carriers. In line with the hypothesis that downregulation of IGF-I signaling is associated with increased dementia risk, ApoE-ε4 homozygotes without prevalent dementia displayed lower levels of IGF-I receptor stimulating activity than heterozygotes and non-carriers. Conclusion: The findings shed new light on the association between IGF-I signaling and the neuropathology of dementia and ask for replication in other cohorts, using measures of IGF-I receptor stimulating activity rather than total serum levels as putative markers of dementia risk.
Collapse
Affiliation(s)
- Sara A Galle
- Department of Clinical, Neuro- and Developmental Psychology, Vrije Universiteit Amsterdam, Amsterdam, Netherlands.,Department of Genetic Epidemiology, Erasmus Medical Center, Rotterdam, Netherlands
| | - Ashley van der Spek
- Department of Genetic Epidemiology, Erasmus Medical Center, Rotterdam, Netherlands
| | - Madeleine L Drent
- Department of Clinical, Neuro- and Developmental Psychology, Vrije Universiteit Amsterdam, Amsterdam, Netherlands.,Section of Endocrinology, Department of Internal Medicine, Amsterdam University Medical Center, Amsterdam, Netherlands
| | - Michael P Brugts
- Department of Internal Medicine, Ikazia Ziekenhuis, Rotterdam, Netherlands
| | - Erik J A Scherder
- Department of Clinical, Neuro- and Developmental Psychology, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | | | - M Arfan Ikram
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, Netherlands.,Department of Neurology, Erasmus Medical Center, Rotterdam, Netherlands.,Department of Radiology, Erasmus Medical Center, Rotterdam, Netherlands
| | - Cornelia M van Duijn
- Department of Genetic Epidemiology, Erasmus Medical Center, Rotterdam, Netherlands.,Nuffield Department of Population Health, Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
27
|
Alonso R, Pisa D, Fernández-Fernández AM, Carrasco L. Infection of Fungi and Bacteria in Brain Tissue From Elderly Persons and Patients With Alzheimer's Disease. Front Aging Neurosci 2018; 10:159. [PMID: 29881346 PMCID: PMC5976758 DOI: 10.3389/fnagi.2018.00159] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 05/08/2018] [Indexed: 12/17/2022] Open
Abstract
Alzheimer's disease (AD) is the leading cause of dementia in elderly people. The etiology of this disease remains a matter of intensive research in many laboratories. We have advanced the idea that disseminated fungal infection contributes to the etiology of AD. Thus, we have demonstrated that fungal proteins and DNA are present in nervous tissue from AD patients. More recently, we have reported that bacterial infections can accompany these mycoses, suggesting that polymicrobial infections exist in AD brains. In the present study, we have examined fungal and bacterial infection in brain tissue from AD patients and control subjects by immunohistochemistry. In addition, we have documented the fungal and bacterial species in brain regions from AD patients and control subjects by next-generation sequencing (NGS). Our results from the analysis of ten AD patients reveal a variety of fungal and bacterial species, although some were more prominent than others. The fungal genera more prevalent in AD patients were Alternaria, Botrytis, Candida, and Malassezia. We also compared these genera with those found in elderly and younger subjects. One of the most prominent genera in control subjects was Fusarium. Principal component analysis clearly indicated that fungi from frontal cortex samples of AD brains clustered together and differed from those of equivalent control subjects. Regarding bacterial infection, the phylum Proteobacteria was the most prominent in both AD patients and controls, followed by Firmicutes, Actinobacteria, and Bacteroides. At the family level, Burkholderiaceae and Staphylococcaceae exhibited higher percentages in AD brains than in control brains. These findings could be of interest to guide targeted antimicrobial therapy for AD patients. Moreover, the variety of microbial species in each patient may constitute a basis for a better understanding of the evolution and severity of clinical symptoms in each patient.
Collapse
Affiliation(s)
| | | | | | - Luis Carrasco
- Centro de Biología Molecular “Severo Ochoa”, Consejo Superior de Investigaciones Científicas y Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
28
|
Skillbäck T, Lautner R, Mattsson N, Schott JM, Skoog I, Nägga K, Kilander L, Wimo A, Winblad B, Eriksdotter M, Blennow K, Zetterberg H. Apolipoprotein E genotypes and longevity across dementia disorders. Alzheimers Dement 2018; 14:895-901. [PMID: 29548722 DOI: 10.1016/j.jalz.2018.02.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 06/02/2017] [Accepted: 02/07/2018] [Indexed: 10/17/2022]
Abstract
INTRODUCTION The ε4 allele of the apolipoprotein E (APOE) gene is a prominent risk factor for Alzheimer's disease (AD), but its implication in other dementias is less well studied. METHODS We used a data set on 2858 subjects (1098 AD, 260 vascular dementia [VaD], 145 mixed AD and VaD, 90 other dementia diagnoses, and 1265 controls) to examine the association of APOE polymorphisms with clinical dementia diagnoses, biomarker profiles, and longevity. RESULTS The ε4 allele was associated with reduced longevity as ε4 versus ε3 homozygotes lived on average 2.6 years shorter (P = .006). In AD, ε4 carriers lived 1.0 years shorter than noncarriers (P = .028). The ε4 allele was more prevalent in AD, mixed AD and VaD, and VaD patients compared to controls, but not in other dementia disorders. DISCUSSION The APOE ε4 allele is influential in AD but might also be of importance in VaD and in mixed AD and VaD, diseases in which concomitant AD pathology is common.
Collapse
Affiliation(s)
- Tobias Skillbäck
- Department of Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.
| | - Ronald Lautner
- Department of Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Niklas Mattsson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden; Department of Neurology, Skåne University Hospital, Lund, Sweden
| | | | - Ingmar Skoog
- Department of Psychiatry and Neurochemistry, Neuropsychiatric Epidemiology Unit, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Katarina Nägga
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Lena Kilander
- Department of Public Health and Caring Sciences/Geriatrics, Uppsala University, Uppsala, Sweden
| | - Anders Wimo
- Department of Neurobiology, Care Sciences, and Society (NVS), Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Huddinge, Sweden; Centre for Research & Development Uppsala University/County Council of Gävleborg, Gävle, Sweden
| | - Bengt Winblad
- Department of Neurobiology, Care Sciences, and Society (NVS), Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Huddinge, Sweden; Department Geriatric Medicine, Karolinska University Hospital, Huddinge, Sweden
| | - Maria Eriksdotter
- Department of Neurobiology, Care Sciences, and Society (NVS), Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, Huddinge, Sweden; Department Geriatric Medicine, Karolinska University Hospital, Huddinge, Sweden
| | - Kaj Blennow
- Department of Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, UK
| |
Collapse
|
29
|
Geerts H, Spiros A, Roberts P. Impact of amyloid-beta changes on cognitive outcomes in Alzheimer's disease: analysis of clinical trials using a quantitative systems pharmacology model. Alzheimers Res Ther 2018; 10:14. [PMID: 29394903 PMCID: PMC5797372 DOI: 10.1186/s13195-018-0343-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 01/15/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Despite a tremendous amount of information on the role of amyloid in Alzheimer's disease (AD), almost all clinical trials testing this hypothesis have failed to generate clinically relevant cognitive effects. METHODS We present an advanced mechanism-based and biophysically realistic quantitative systems pharmacology computer model of an Alzheimer-type neuronal cortical network that has been calibrated with Alzheimer Disease Assessment Scale, cognitive subscale (ADAS-Cog) readouts from historical clinical trials and simulated the differential impact of amyloid-beta (Aβ40 and Aβ42) oligomers on glutamate and nicotinic neurotransmission. RESULTS Preclinical data suggest a beneficial effect of shorter Aβ forms within a limited dose range. Such a beneficial effect of Aβ40 on glutamate neurotransmission in human patients is absolutely necessary to reproduce clinical data on the ADAS-Cog in minimal cognitive impairment (MCI) patients with and without amyloid load, the effect of APOE genotype effect on the slope of the cognitive trajectory over time in placebo AD patients and higher sensitivity to cholinergic manipulation with scopolamine associated with higher Aβ in MCI subjects. We further derive a relationship between units of Aβ load in our model and the standard uptake value ratio from amyloid imaging. When introducing the documented clinical pharmacodynamic effects on Aβ levels for various amyloid-related clinical interventions in patients with low Aβ baseline, the platform predicts an overall significant worsening for passive vaccination with solanezumab, beta-secretase inhibitor verubecestat and gamma-secretase inhibitor semagacestat. In contrast, all three interventions improved cognition in subjects with moderate to high baseline Aβ levels, with verubecestat anticipated to have the greatest effect (around ADAS-Cog value 1.5 points), solanezumab the lowest (0.8 ADAS-Cog value points) and semagacestat in between. This could explain the success of many amyloid interventions in transgene animals with an artificial high level of Aβ, but not in AD patients with a large variability of amyloid loads. CONCLUSIONS If these predictions are confirmed in post-hoc analyses of failed clinical amyloid-modulating trials, one should question the rationale behind testing these interventions in early and prodromal subjects with low or zero amyloid load.
Collapse
Affiliation(s)
- Hugo Geerts
- In Silico Biosciences, 686 Westwind Dr, Berwyn, PA, 1312, USA.
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Athan Spiros
- In Silico Biosciences, 686 Westwind Dr, Berwyn, PA, 1312, USA
| | - Patrick Roberts
- In Silico Biosciences, 686 Westwind Dr, Berwyn, PA, 1312, USA
- Amazon AI AWS, Portland, OR, USA
| |
Collapse
|
30
|
Lin S, Liu X, Yao B, Huang Z. Controlling for confounding factors and revealing their interactions in genetic association meta-analyses: a computing method and application for stratification analyses. Oncotarget 2018; 9:12125-12136. [PMID: 29552297 PMCID: PMC5844733 DOI: 10.18632/oncotarget.24335] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 01/24/2018] [Indexed: 11/25/2022] Open
Abstract
Subgroup and stratification analyses have been widely applied in genetic association studies to compare the effects of different factors or control for the effects of the confounding variables associated with a disease. However, studies have not systematically provided application standards and computing methods for stratification analyses. Based on the Mantel-Haenszel and Inverse-Variant approaches and two practical computing methods described in previous studies, we propose a standard stratification method for meta-analyses that contains two sequential steps: factorial stratification analysis and confounder-controlling stratification analysis. Examples of genetic association meta-analyses are used to illustrate these points. The standard stratification analysis method identifies interacting effects on investigated factors and controls for confounding variables, and this method effectively reveals the real effects of these factors and confounding variables on a disease in an overall study population. We also discuss important issues concerning stratification for meta-analyses, such as conceptual confusion between subgroup and stratification analyses, and incorrect calculations previously used for factorial stratification analyses. This standard stratification method will have extensive applications in future research for increasing studies on the complicated relationships between genetics and disease.
Collapse
Affiliation(s)
- Shuhuang Lin
- Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, Guangdong 523808, China.,The Second School of Clinical Medicine, Guangdong Medical University, Dongguan, Guangdong 523808, China
| | - Xu Liu
- Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, Guangdong 523808, China.,The Second School of Clinical Medicine, Guangdong Medical University, Dongguan, Guangdong 523808, China
| | - Bin Yao
- Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, Guangdong 523808, China.,The Second School of Clinical Medicine, Guangdong Medical University, Dongguan, Guangdong 523808, China
| | - Zunnan Huang
- Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Dongguan Scientific Research Center, Guangdong Medical University, Dongguan, Guangdong 523808, China.,Institute of Marine Biomedical Research, Guangdong Medical University, Zhanjiang, Guangdong 524023, China
| |
Collapse
|
31
|
Marottoli FM, Katsumata Y, Koster KP, Thomas R, Fardo DW, Tai LM. Peripheral Inflammation, Apolipoprotein E4, and Amyloid-β Interact to Induce Cognitive and Cerebrovascular Dysfunction. ASN Neuro 2017; 9:1759091417719201. [PMID: 28707482 PMCID: PMC5521356 DOI: 10.1177/1759091417719201] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cerebrovascular dysfunction is rapidly reemerging as a major process of Alzheimer’s disease (AD). It is, therefore, crucial to delineate the roles of AD risk factors in cerebrovascular dysfunction. While apolipoprotein E4 (APOE4), Amyloid-β (Aβ), and peripheral inflammation independently induce cerebrovascular damage, their collective effects remain to be elucidated. The goal of this study was to determine the interactive effect of APOE4, Aβ, and chronic repeated peripheral inflammation on cerebrovascular and cognitive dysfunction in vivo. EFAD mice are a well-characterized mouse model that express human APOE3 (E3FAD) or APOE4 (E4FAD) and overproduce human Aβ42 via expression of 5 Familial Alzheimer’s disease (5xFAD) mutations. Here, we utilized EFAD carriers [5xFAD+/−/APOE+/+ (EFAD+)] and noncarriers [5xFAD−/−/APOE+/+ (EFAD−)] to compare the effects of peripheral inflammation in the presence or absence of human Aβ overproduction. Low-level, chronic repeated peripheral inflammation was induced in EFAD mice via systemic administration of lipopolysaccharide (LPS; 0.5 mg/kg/wk i.p.) from 4 to 6 months of age. In E4FAD+ mice, peripheral inflammation caused cognitive deficits and lowered post-synaptic protein levels. Importantly, cerebrovascular deficits were observed in LPS-challenged E4FAD+ mice, including cerebrovascular leakiness, lower vessel coverage, and cerebral amyloid angiopathy-like Aβ deposition. Thus, APOE4, Aβ, and peripheral inflammation interact to induce cerebrovascular damage and cognitive deficits.
Collapse
Affiliation(s)
- Felecia M Marottoli
- 1 Department of Anatomy and Cell Biology, University of Illinois at Chicago, IL, USA
| | - Yuriko Katsumata
- 2 Department of Biostatistics, University of Kentucky, Lexington, KY, USA
| | - Kevin P Koster
- 1 Department of Anatomy and Cell Biology, University of Illinois at Chicago, IL, USA
| | - Riya Thomas
- 1 Department of Anatomy and Cell Biology, University of Illinois at Chicago, IL, USA
| | - David W Fardo
- 2 Department of Biostatistics, University of Kentucky, Lexington, KY, USA.,3 Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Leon M Tai
- 1 Department of Anatomy and Cell Biology, University of Illinois at Chicago, IL, USA
| |
Collapse
|
32
|
Tai LM, Balu D, Avila-Munoz E, Abdullah L, Thomas R, Collins N, Valencia-Olvera AC, LaDu MJ. EFAD transgenic mice as a human APOE relevant preclinical model of Alzheimer's disease. J Lipid Res 2017; 58:1733-1755. [PMID: 28389477 PMCID: PMC5580905 DOI: 10.1194/jlr.r076315] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 04/06/2017] [Indexed: 01/12/2023] Open
Abstract
Identified in 1993, APOE4 is the greatest genetic risk factor for sporadic Alzheimer's disease (AD), increasing risk up to 15-fold compared with APOE3, with APOE2 decreasing AD risk. However, the functional effects of APOE4 on AD pathology remain unclear and, in some cases, controversial. In vivo progress to understand how the human (h)-APOE genotypes affect AD pathology has been limited by the lack of a tractable familial AD-transgenic (FAD-Tg) mouse model expressing h-APOE rather than mouse (m)-APOE. The disparity between m- and h-apoE is relevant for virtually every AD-relevant pathway, including amyloid-β (Aβ) deposition and clearance, neuroinflammation, tau pathology, neural plasticity and cerebrovascular deficits. EFAD mice were designed as a temporally useful preclinical FAD-Tg-mouse model expressing the h-APOE genotypes for identifying mechanisms underlying APOE-modulated symptoms of AD pathology. From their first description in 2012, EFAD mice have enabled critical basic and therapeutic research. Here we review insights gleaned from the EFAD mice and summarize future directions.
Collapse
Affiliation(s)
- Leon M Tai
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612
| | - Deebika Balu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612
| | - Evangelina Avila-Munoz
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612
| | | | - Riya Thomas
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612
| | - Nicole Collins
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612
| | | | - Mary Jo LaDu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612.
| |
Collapse
|
33
|
Huynh RA, Mohan C. Alzheimer's Disease: Biomarkers in the Genome, Blood, and Cerebrospinal Fluid. Front Neurol 2017; 8:102. [PMID: 28373857 PMCID: PMC5357660 DOI: 10.3389/fneur.2017.00102] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 03/01/2017] [Indexed: 01/20/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that slowly destroys memory and thinking skills, resulting in behavioral changes. It is estimated that nearly 36 million are affected globally with numbers reaching 115 million by 2050. AD can only be definitively diagnosed at autopsy since its manifestations of senile plaques and neurofibrillary tangles throughout the brain cannot yet be fully captured with current imaging technologies. Current AD therapeutics have also been suboptimal. Besides identifying markers that distinguish AD from controls, there has been a recent drive to identify better biomarkers that can predict the rates of cognitive decline and neocortical amyloid burden in those who exhibit preclinical, prodromal, or clinical AD. This review covers biomarkers of three main types: genes, cerebrospinal fluid-derived, and blood-derived biomarkers. Looking ahead, cutting-edge OMICs technologies, including proteomics and metabolomics, ought to be fully tapped in order to mine even better biomarkers for AD that are more predictive.
Collapse
Affiliation(s)
- Rose Ann Huynh
- Department of Biomedical Engineering, University of Houston , Houston, TX , USA
| | - Chandra Mohan
- Department of Biomedical Engineering, University of Houston , Houston, TX , USA
| |
Collapse
|
34
|
Counts SE, Ikonomovic MD, Mercado N, Vega IE, Mufson EJ. Biomarkers for the Early Detection and Progression of Alzheimer's Disease. Neurotherapeutics 2017; 14:35-53. [PMID: 27738903 PMCID: PMC5233625 DOI: 10.1007/s13311-016-0481-z] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The recent failures of potential disease-modifying drugs for Alzheimer's disease (AD) may reflect the fact that the enrolled participants in clinical trials are already too advanced to derive a clinical benefit. Thus, well-validated biomarkers for the early detection and accurate diagnosis of the preclinical stages of AD will be crucial for therapeutic advancement. The combinatorial use of biomarkers derived from biological fluids, such as cerebrospinal fluid (CSF), with advanced molecular imaging and neuropsychological testing may eventually achieve the diagnostic sensitivity and specificity necessary to identify people in the earliest stages of the disease when drug modification is most likely possible. In this regard, positive amyloid or tau tracer retention on positron emission tomography imaging, low CSF concentrations of the amyloid-β 1-42 peptide, high CSF concentrations in total tau and phospho-tau, mesial temporal lobe atrophy on magnetic resonance imaging, and temporoparietal/precuneus hypometabolism or hypoperfusion on 18F-fluorodeoxyglucose positron emission tomography have all emerged as biomarkers for the progression to AD. However, the ultimate AD biomarker panel will likely involve the inclusion of novel CSF and blood biomarkers more precisely associated with confirmed pathophysiologic mechanisms to improve its reliability for detecting preclinical AD. This review highlights advancements in biological fluid and imaging biomarkers that are moving the field towards achieving the goal of a preclinical detection of AD.
Collapse
Affiliation(s)
- Scott E Counts
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
- Department of Family Medicine, Michigan State University, Grand Rapids, MI, USA
- Hauenstein Neuroscience Center, Mercy Health Saint Mary's Hospital, Grand Rapids, MI, USA
| | - Milos D Ikonomovic
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, PA, USA
| | - Natosha Mercado
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Irving E Vega
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Elliott J Mufson
- Department of Neurobiology and Neurology, Barrow Neurological Institute, Phoenix, AZ, USA.
| |
Collapse
|
35
|
Jiang T, Sun Q, Chen S. Oxidative stress: A major pathogenesis and potential therapeutic target of antioxidative agents in Parkinson's disease and Alzheimer's disease. Prog Neurobiol 2016; 147:1-19. [PMID: 27769868 DOI: 10.1016/j.pneurobio.2016.07.005] [Citation(s) in RCA: 425] [Impact Index Per Article: 53.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 06/19/2016] [Accepted: 07/11/2016] [Indexed: 12/14/2022]
Abstract
Oxidative stress reflects an imbalance between the overproduction and incorporation of free radicals and the dynamic ability of a biosystem to detoxify reactive intermediates. Free radicals produced by oxidative stress are one of the common features in several experimental models of diseases. Free radicals affect both the structure and function of neural cells, and contribute to a wide range of neurodegenerative diseases, including Parkinson's disease and Alzheimer's disease. Although the precise mechanisms that result in the degeneration of neurons and the relevant pathological changes remain unclear, the crucial role of oxidative stress in the pathogenesis of neurodegenerative diseases is associated with several proteins (such as α-synuclein, DJ-1, Amyloid β and tau protein) and some signaling pathways (such as extracellular regulated protein kinases, phosphoinositide 3-kinase/Protein Kinase B pathway and extracellular signal-regulated kinases 1/2) that are tightly associated with the neural damage. In this review, we present evidence, gathered over the last decade, concerning a variety of pathogenic proteins, their important signaling pathways and pathogenic mechanisms associated with oxidative stress in Parkinson's disease and Alzheimer's disease. Proper control and regulation of these proteins' functions and the related signaling pathways may be a promising therapeutic approach to the patients. We also emphasizes antioxidative options, including some new neuroprotective agents that eliminate excess reactive oxygen species efficiently and have a certain therapeutic effect; however, controversy surrounds some of them in terms of the dose and length of therapy. These agents require further investigation by clinical application in patients suffering Parkinson's disease and Alzheimer's disease.
Collapse
Affiliation(s)
- Tianfang Jiang
- Department of Neurology, Institute of Neurology and the Collaborative Innovation Center for Brain Science, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Qian Sun
- Department of Neurology, Institute of Neurology and the Collaborative Innovation Center for Brain Science, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Shengdi Chen
- Department of Neurology, Institute of Neurology and the Collaborative Innovation Center for Brain Science, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Laboratory of Neurodegenerative Diseases, Institute of Health Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Science & Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
36
|
A systematic review and meta-analysis of the association between the apolipoprotein E genotype and delirium. Psychiatr Genet 2016; 26:53-9. [PMID: 26901792 DOI: 10.1097/ypg.0000000000000122] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The role of apolipoprotein E (APOE) in Alzheimer's disease and other dementias has been investigated intensively. However, the relationship between APOE and delirium has only recently been explored in studies that have included relatively small samples. A meta-analysis of the published pooled data is timely to explore the relationship between APOE and delirium and to inform further research in this topic. PubMed, EBSCOhost, Google Scholar, Scopus, all EBM Reviews (OVID) and the Cochrane Database of Systematic Reviews were searched with relevant keywords and from the references of relevant papers. Ten papers were found that examined the relationship between APOE and delirium. Data were extracted from eight of them and pooled for meta-analysis using random effects with R software. Data from 1762 participants, of whom 479 (27.2%) were diagnosed with delirium, showed low heterogeneity (Q=13.11, d.f.=7, P=0.07; I=44.86%). The possession of the APOE ε4 allele has a small (log odds ratio: 0.18, 95% confidence interval: 0.23-0.59), nonsignificant (P=0.38) effect on the presence of delirium. No publication bias was identified. The metapower of the pooled data was low (α=0.05, power=0.65). On analysing the studies to date, it seems that there is no association between APOE and the occurrence of delirium. We suggest that further studies are needed with greater number of patients to clarify any association as well as to examine for other patterns of association including relevance for subgroups of patients who develop delirium and for effects on the phenotype of delirium and the outcomes.
Collapse
|
37
|
Influence of APOE Genotype on Alzheimer's Disease CSF Biomarkers in a Spanish Population. BIOMED RESEARCH INTERNATIONAL 2016; 2016:1390620. [PMID: 27092308 PMCID: PMC4820585 DOI: 10.1155/2016/1390620] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 10/09/2015] [Accepted: 11/25/2015] [Indexed: 11/18/2022]
Abstract
Objectives. To evaluate the association between apolipoprotein E (APOE) genotype and cerebrospinal fluid (CSF) levels of Alzheimer's disease (AD) biomarkers and to study the influence of APOE genotype on the development of AD in a Spanish population. Material and Methods. The study comprised 29 amnestic mild cognitive impairment (MCI) patients and 27 control subjects. Using ELISA methodology, CSF biomarkers and tau/Aβ ratios were obtained. ANOVA and adjusted odds ratios were calculated. Results. We observed the effect of APOE genotype and age on CSF AD variables. The progression to AD was more clearly influenced by CSF AD variables than by age or APOE status. Conclusions. APOE status influences CSF AD variables. However, the presence of APOE ε4 does not appear to be a deterministic factor for the development of AD, because CSF variables have a greater influence on progression to the disease. These results confirm previous observations and, to our knowledge, are the first published in a Spanish population.
Collapse
|
38
|
Dubois B, Hampel H, Feldman HH, Scheltens P, Aisen P, Andrieu S, Bakardjian H, Benali H, Bertram L, Blennow K, Broich K, Cavedo E, Crutch S, Dartigues JF, Duyckaerts C, Epelbaum S, Frisoni GB, Gauthier S, Genthon R, Gouw AA, Habert MO, Holtzman DM, Kivipelto M, Lista S, Molinuevo JL, O'Bryant SE, Rabinovici GD, Rowe C, Salloway S, Schneider LS, Sperling R, Teichmann M, Carrillo MC, Cummings J, Jack CR. Preclinical Alzheimer's disease: Definition, natural history, and diagnostic criteria. Alzheimers Dement 2016; 12:292-323. [PMID: 27012484 PMCID: PMC6417794 DOI: 10.1016/j.jalz.2016.02.002] [Citation(s) in RCA: 1197] [Impact Index Per Article: 149.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
During the past decade, a conceptual shift occurred in the field of Alzheimer's disease (AD) considering the disease as a continuum. Thanks to evolving biomarker research and substantial discoveries, it is now possible to identify the disease even at the preclinical stage before the occurrence of the first clinical symptoms. This preclinical stage of AD has become a major research focus as the field postulates that early intervention may offer the best chance of therapeutic success. To date, very little evidence is established on this "silent" stage of the disease. A clarification is needed about the definitions and lexicon, the limits, the natural history, the markers of progression, and the ethical consequence of detecting the disease at this asymptomatic stage. This article is aimed at addressing all the different issues by providing for each of them an updated review of the literature and evidence, with practical recommendations.
Collapse
Affiliation(s)
- Bruno Dubois
- Institute of Memory and Alzheimer's Disease (IM2A) and Brain and Spine Institute (ICM) UMR S 1127 Frontlab, Department of Neurology, AP_HP, Pitié-Salpêtrière University Hospital, Sorbonne Universities, Pierre et Marie Curie University, Paris 06, Paris, France.
| | - Harald Hampel
- Institute of Memory and Alzheimer's Disease (IM2A) and Brain and Spine Institute (ICM) UMR S 1127 Frontlab, Department of Neurology, AP_HP, Pitié-Salpêtrière University Hospital, Sorbonne Universities, Pierre et Marie Curie University, Paris 06, Paris, France; AXA Research Fund & UPMC Chair, Paris, France
| | | | - Philip Scheltens
- Department of Neurology and Alzheimer Center, VU University Medical Center and Neuroscience Campus, Amsterdam, The Netherlands
| | - Paul Aisen
- University of Southern California San Diego, CA, USA
| | - Sandrine Andrieu
- UMR1027, INSERM, Université Toulouse III, Toulouse University Hospital, France
| | - Hovagim Bakardjian
- IHU-A-ICM-Institut des Neurosciences translationnelles de Paris, Paris, France
| | - Habib Benali
- INSERM U1146-CNRS UMR 7371-UPMC UM CR2, Site Pitié-Salpêtrière, Paris, France
| | - Lars Bertram
- Lübeck Interdisciplinary Platform for Genome Analytics (LIGA), Institutes of Neurogenetics and Integrative and Experimental Genomics, University of Lübeck, Lübeck, Germany; School of Public Health, Faculty of Medicine, Imperial College London, London, UK
| | - Kaj Blennow
- Clinical Neurochemistry Lab, Department of Neuroscience and Physiology, University of Gothenburg, Mölndal Hospital, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Karl Broich
- Federal Institute for Drugs and Medical Devices, Bonn, Germany
| | - Enrica Cavedo
- AXA Research Fund & UPMC Chair, Paris, France; Laboratory of Alzheimer's Neuroimaging and Epidemiology, IRCCS Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Sebastian Crutch
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Institute of Neurology, University College London, London, UK
| | | | - Charles Duyckaerts
- University Pierre et Marie Curie, Assistance Publique des Hôpitaux de Paris, Alzheimer-Prion Team Institut du Cerveau et de la Moelle (ICM), Paris, France
| | - Stéphane Epelbaum
- Institute of Memory and Alzheimer's Disease (IM2A) and Brain and Spine Institute (ICM) UMR S 1127 Frontlab, Department of Neurology, AP_HP, Pitié-Salpêtrière University Hospital, Sorbonne Universities, Pierre et Marie Curie University, Paris 06, Paris, France
| | - Giovanni B Frisoni
- University Hospitals and University of Geneva, Geneva, Switzerland; IRCCS Fatebenefratelli, Brescia, Italy
| | - Serge Gauthier
- McGill Center for Studies in Aging, Douglas Mental Health Research Institute, Montreal, Canada
| | - Remy Genthon
- Fondation pour la Recherche sur Alzheimer, Hôpital Pitié-Salpêtrière, Paris, France
| | - Alida A Gouw
- UMR1027, INSERM, Université Toulouse III, Toulouse University Hospital, France; Department of Clinical Neurophysiology/MEG Center, VU University Medical Center, Amsterdam
| | - Marie-Odile Habert
- Sorbonne Universités, UPMC Univ Paris 06, CNRS, INSERM, Laboratoire d'Imagerie Biomédicale, Paris, France; AP-HP, Hôpital Pitié-Salpêtrière, Département de Médecine Nucléaire, Paris, France
| | - David M Holtzman
- Department of Neurology, Washington University, Hope Center for Neurological Disorders, St. Louis, MO, USA; Department of Neurology, Washington University, Knight Alzheimer's Disease Research Center, St. Louis, MO, USA
| | - Miia Kivipelto
- Center for Alzheimer Research, Karolinska Institutet, Department of Geriatric Medicine, Karolinska University Hospital, Stockholm, Sweden; Institute of Clinical Medicine/ Neurology, University of Eastern Finland, Kuopio, Finland
| | | | - José-Luis Molinuevo
- Alzheimer's Disease and Other Cognitive Disorders Unit, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain
| | - Sid E O'Bryant
- Center for Alzheimer's & Neurodegenerative Disease Research, University of North Texas Health Science Center, TX, USA
| | - Gil D Rabinovici
- Memory & Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Christopher Rowe
- Department of Molecular Imaging, Austin Health, University of Melbourne, Australia
| | - Stephen Salloway
- Memory and Aging Program, Butler Hospital, Alpert Medical School of Brown University, USA; Department of Neurology, Alpert Medical School of Brown University, USA; Department of Psychiatry, Alpert Medical School of Brown University, USA
| | - Lon S Schneider
- Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Reisa Sperling
- Harvard Medical School, Memory Disorders Unit, Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Boston, USA; Harvard Medical School, Memory Disorders Unit, Center for Alzheimer Research and Treatment, Massachusetts General Hospital, Boston, USA
| | - Marc Teichmann
- Institute of Memory and Alzheimer's Disease (IM2A) and Brain and Spine Institute (ICM) UMR S 1127 Frontlab, Department of Neurology, AP_HP, Pitié-Salpêtrière University Hospital, Sorbonne Universities, Pierre et Marie Curie University, Paris 06, Paris, France
| | - Maria C Carrillo
- The Alzheimer's Association Division of Medical & Scientific Relations, Chicago, USA
| | - Jeffrey Cummings
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
| | - Cliff R Jack
- Department of Radiology, Mayo Clinic, Rochester MN, USA
| |
Collapse
|
39
|
Orellana C, Ferreira D, Muehlboeck JS, Mecocci P, Vellas B, Tsolaki M, Kłoszewska I, Soininen H, Lovestone S, Simmons A, Wahlund LO, Westman E. Measuring Global Brain Atrophy with the Brain Volume/Cerebrospinal Fluid Index: Normative Values, Cut-Offs and Clinical Associations. NEURODEGENER DIS 2015; 16:77-86. [PMID: 26726737 DOI: 10.1159/000442443] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 11/11/2015] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Global brain atrophy is present in normal aging and different neurodegenerative disorders such as Alzheimer's disease (AD) and is becoming widely used to monitor disease progression. SUMMARY The brain volume/cerebrospinal fluid index (BV/CSF index) is validated in this study as a measurement of global brain atrophy. We tested the ability of the BV/CSF index to detect global brain atrophy, investigated the influence of confounders, provided normative values and cut-offs for mild, moderate and severe brain atrophy, and studied associations with different outcome variables. A total of 1,009 individuals were included [324 healthy controls, 408 patients with mild cognitive impairment (MCI) and 277 patients with AD]. Magnetic resonance images were segmented using FreeSurfer, and the BV/CSF index was calculated and studied both cross-sectionally and longitudinally (1-year follow-up). Both AD patients and MCI patients who progressed to AD showed greater global brain atrophy compared to stable MCI patients and controls. Atrophy was associated with older age, larger intracranial volume, less education and presence of the ApoE ε4 allele. Significant correlations were found with clinical variables, CSF biomarkers and several cognitive tests. KEY MESSAGES The BV/CSF index may be useful for staging individuals according to the degree of global brain atrophy, and for monitoring disease progression. It also shows potential for predicting clinical changes and for being used in the clinical routine.
Collapse
|
40
|
DeCarlo CA, MacDonald SWS, Vergote D, Jhamandas J, Westaway D, Dixon RA. Vascular Health and Genetic Risk Affect Mild Cognitive Impairment Status and 4-Year Stability: Evidence From the Victoria Longitudinal Study. J Gerontol B Psychol Sci Soc Sci 2015; 71:1004-1014. [PMID: 26362601 DOI: 10.1093/geronb/gbv043] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Accepted: 05/16/2015] [Indexed: 01/12/2023] Open
Abstract
OBJECTIVES Mild cognitive impairment (MCI) is a high-risk condition for progression to Alzheimer's disease (AD). Vascular health is a key mechanism underlying age-related cognitive decline and neurodegeneration. AD-related genetic risk factors may be associated with preclinical cognitive status changes. We examine independent and cross-domain interactive effects of vascular and genetic markers for predicting MCI status and stability. METHOD We used cross-sectional and 2-wave longitudinal data from the Victoria Longitudinal Study, including indicators of vascular health (e.g., reported vascular diseases, measured lung capacity and pulse rate) and genetic risk factors-that is, apolipoprotein E (APOE; rs429358 and rs7412; the presence vs absence of ε4) and catechol-O-methyltransferase (COMT; rs4680; met/met vs val/val). We examined associations with objectively classified (a) cognitive status at baseline (not impaired congnitive (NIC) controls vs MCI) and (b) stability or transition of cognitive status across a 4-year interval (stable NIC-NIC vs chronic MCI-MCI or transitional NIC-MCI). RESULTS Using logistic regression, indicators of vascular health, both independently and interactively with APOE ε4, were associated with risk of MCI at baseline and/or associated with MCI conversion or MCI stability over the retest interval. DISCUSSION Several vascular health markers of aging predict MCI risk. Interactively, APOE ε4 may intensify the vascular health risk for MCI.
Collapse
Affiliation(s)
- Correne A DeCarlo
- Department of Psychology, University of Victoria, British Columbia, Canada.
| | | | | | - Jack Jhamandas
- Department of Medicine (Neurology).,Neuroscience and Mental Health Institute
| | - David Westaway
- Neuroscience and Mental Health Institute.,Centre for Prions and Protein Folding Diseases
| | - Roger A Dixon
- Neuroscience and Mental Health Institute.,Department of Psychology, University of Alberta, Edmonton, Canada
| |
Collapse
|
41
|
Hua R, Wei M, Zhang C. The complex genetics in autism spectrum disorders. SCIENCE CHINA-LIFE SCIENCES 2015; 58:933-45. [PMID: 26335739 DOI: 10.1007/s11427-015-4893-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Autism spectrum disorders (ASD) are a pervasive neurodevelopmental disease characterized by deficits in social interaction and nonverbal communication, as well as restricted interests and stereotypical behavior. Genetic changes/heritability is one of the major contributing factors, and hundreds to thousands of causative and susceptible genes, copy number variants (CNVs), linkage regions, and microRNAs have been associated with ASD which clearly indicates that ASD is a complex genetic disorder. Here, we will briefly summarize some of the high-confidence genetic changes in ASD and their possible roles in their pathogenesis.
Collapse
Affiliation(s)
- Rui Hua
- State Key Laboratory of Membrane Biology, School of Life Sciences; PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - MengPing Wei
- State Key Laboratory of Membrane Biology, School of Life Sciences; PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Chen Zhang
- State Key Laboratory of Membrane Biology, School of Life Sciences; PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China.
| |
Collapse
|
42
|
Liu S, Zeng F, Wang C, Chen Z, Zhao B, Li K. The nitric oxide synthase 3 G894T polymorphism associated with Alzheimer's disease risk: a meta-analysis. Sci Rep 2015; 5:13598. [PMID: 26337484 PMCID: PMC4559797 DOI: 10.1038/srep13598] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 07/27/2015] [Indexed: 12/23/2022] Open
Abstract
The association between the G894T polymorphism (Glu298Asp) of nitric oxide synthase 3 (NOS3) and risk of Alzheimer’s disease (AD) was explored by performing a meta-analysis of case-control studies. Bibliographical searches were conducted in the MEDLINE, EMBASE, and China National Knowledge Infrastructure (CNKI) databases without any language limitations. Two investigators independently assessed abstracts for relevant studies, and reviewed all eligible studies. We adopted regrouping in accordance with the most probably appropriate genetic model. Odds ratios (ORs) with 95% confidence intervals (CIs) were used to assess the strength of this association. We performed a meta-analysis including 21 published articles with 23 case-control studies (5,670 cases and 5,046 controls). In the analyses, we found significant association between G894T polymorphism and AD risk under a complete overdominant model (GG + TT vs. GT) (OR = 1.18; 95%CI, 1.04–1.35; P = 0.010). When stratified by time of AD onset, we found the association between this polymorphism and AD susceptibility to be more substantial among late onset patients than among early onset patients (OR for late vs. early onset: 1.33 vs. 1.02, P interaction = 0.049). The meta-analysis showed that the polymorphism G894T of NOS3 was associated with risk of AD.
Collapse
Affiliation(s)
- Shengyuan Liu
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical College, Zhanjiang 524001, China.,Department of Chronic Disease, Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen 518054, China
| | - Fangfang Zeng
- Epidemiology research unit, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Changyi Wang
- Department of Chronic Disease, Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen 518054, China
| | - Zhongwei Chen
- Department of Chronic Disease, Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen 518054, China
| | - Bin Zhao
- Institute of Neurology, Guangdong Medical College, Zhanjiang 524001, China
| | - Keshen Li
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical College, Zhanjiang 524001, China
| |
Collapse
|
43
|
Sun S, Fu J, Chen J, Pang W, Hu R, Li H, Tan L, Jiang Y. ApoE type 4 allele affects cognitive function of aged population in Tianjin City, China. Am J Alzheimers Dis Other Demen 2015; 30:503-7. [PMID: 25585996 PMCID: PMC10852675 DOI: 10.1177/1533317514566114] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Apolipoprotein E type 4 allele (ApoE∊4) is known as a risk gene for the late-onset Alzheimer's disease, and the relationship between ApoE∊4 and cognitive function of the elderly people has drawn the attention of the scientists. In this study, we investigated the relationship between ApoE∊4 and the cognitive function of the old people. A total of 156 old people were investigated, of whom 31 were ApoE∊4 carriers. The ApoE∊4 primarily influenced the global cognitive function, perceptual speed, and work memory. The results indicate that ApoE∊4 has significant negative effect on the cognitive function of the elderly people who are 60 years and older.
Collapse
Affiliation(s)
- Shoudan Sun
- Department of Nutrition, Tianjin Institute of Health & Environmental Medicine, Tianjin, China Department of Nutrition, Tianjin Hospital, Tianjin, China
| | - Jingming Fu
- Department of Nutrition, Tianjin Institute of Health & Environmental Medicine, Tianjin, China
| | - Jun Chen
- College of Acupuncture and tuina, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wei Pang
- Department of Nutrition, Tianjin Institute of Health & Environmental Medicine, Tianjin, China
| | - Ruomei Hu
- Department of Nutrition, Tianjin Hospital, Tianjin, China
| | - Haiqiang Li
- Department of Nutrition, Tianjin Institute of Health & Environmental Medicine, Tianjin, China
| | - Long Tan
- Department of Nutrition, Tianjin Institute of Health & Environmental Medicine, Tianjin, China
| | - Yugang Jiang
- Department of Nutrition, Tianjin Institute of Health & Environmental Medicine, Tianjin, China
| |
Collapse
|
44
|
Liu SY, Zhao HD, Wang JL, Huang T, Tian HW, Yao LF, Tao H, Chen ZW, Wang CY, Sheng ST, Li H, Zhao B, Li KS. Association between Polymorphisms of the AKT1 Gene Promoter and Risk of the Alzheimer's Disease in a Chinese Han Population with Type 2 Diabetes. CNS Neurosci Ther 2015; 21:619-25. [PMID: 26178916 DOI: 10.1111/cns.12430] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 05/25/2015] [Accepted: 06/08/2015] [Indexed: 12/17/2022] Open
Abstract
AIMS Alzheimer's disease (AD) is a multifactor disease that has been reported to have a close association with type 2 diabetes (T2D) where the v-akt murine thymoma viral oncogene homolog 1 (AKT1) plays an important role in the protein synthesis pathways and cell apoptosis processes. Evidence has been shown that AKT1 protein may be related to AD risk among patients with T2D. The aim of this study was to analyze the potential association between single nucleotide polymorphisms of AKT1 promoter and the risk of AD among patients with T2D. METHODS The association between AKT1 polymorphisms and AD risk in patients with T2D was assessed among 574 consecutive unrelated subjects including 112 AD patients with T2D, 231 patients with AD, and 231 healthy controls in a case-control study. The cognitive function of all subjects was assessed using MMSE. Six single nucleotide polymorphisms with minor allele frequency >0.2 (rs2498786, rs74090038, rs2494750, rs2494751, rs5811155, and rs2494752) in AKT1 promoter were analyzed by polymerase chain reaction (PCR), and the concentration of AKT1 protein in serum was tested using enzyme-linked immunosorbent assay (ELISA). RESULTS Overall, there was statistically significant difference in AKT1 rs2498786 polymorphism. The CC frequency of AKT1 rs2498786 polymorphism in AD with T2D group and AD control group was significantly higher than that in healthy control group (PAD+T2D vs. health < 0.0001, PAD vs. health < 0.0001). However, the difference was not found between AD with T2D group and AD control group. Compared with healthy control group, the plasma levels of AKT1 protein in AD with T2D group (PAD+T2D vs. health < 0.0001) and AD control group (PAD vs. health = 0.0003) decreased significantly. Among genotypes of AKT1 rs2498786 polymorphism, the AKT1 protein level in GG genotype was significantly higher than that in GC genotype (PGG vs. GC < 0.0001) and CC genotype (PGG vs. CC < 0.0001). CONCLUSION The study suggests that AKT1 rs2498786 polymorphism in insulin signaling pathway may be associated with AD risk and different genotypes may affects levels of protein expression. However, the polymorphism is not shown to be exclusive in AD patients with T2D.
Collapse
Affiliation(s)
- Sheng-Yuan Liu
- Key Laboratory of Aging-related cardio-cerebral Diseases of Guangdong Province, Affiliated Hospital of Guangdong Medical College, Zhanjiang, China.,Department of Chronic Noncommunicable Disease Control, Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen, China
| | - He-Dan Zhao
- Department of Clinical Psychology, The Kangci Hospital, Jiaxing, China
| | - Jin-Long Wang
- Department of Clinical Psychology, The Kangci Hospital, Jiaxing, China
| | - Tong Huang
- Community Health Service Center of Nanyou, Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen, China
| | - Hua-Wei Tian
- Department of Prevention and Health, The Futian People's Hospital, Shenzhen, China
| | - Li-Fen Yao
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hua Tao
- Key Laboratory of Aging-related cardio-cerebral Diseases of Guangdong Province, Affiliated Hospital of Guangdong Medical College, Zhanjiang, China
| | - Zhong-Wei Chen
- Department of Chronic Noncommunicable Disease Control, Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen, China
| | - Chang-Yi Wang
- Department of Chronic Noncommunicable Disease Control, Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen, China
| | - Si-Tong Sheng
- College of Life Sciences, Shenzhen University, Shenzhen, China
| | - Hua Li
- HYK High-throughput Biotechnology Institute, Shenzhen, China
| | - Bin Zhao
- Institute of Neurology, Guangdong Medical College, Zhanjiang, China
| | - Ke-Shen Li
- Key Laboratory of Aging-related cardio-cerebral Diseases of Guangdong Province, Affiliated Hospital of Guangdong Medical College, Zhanjiang, China
| |
Collapse
|
45
|
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: 7.9] [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
|
46
|
Gupta VB, Wilson AC, Burnham S, Hone E, Pedrini S, Laws SM, Lim WLF, Rembach A, Rainey-Smith S, Ames D, Cobiac L, Macaulay SL, Masters CL, Rowe CC, Bush AI, Martins RN. Follow-up plasma apolipoprotein E levels in the Australian Imaging, Biomarkers and Lifestyle Flagship Study of Ageing (AIBL) cohort. ALZHEIMERS RESEARCH & THERAPY 2015; 7:16. [PMID: 25859282 PMCID: PMC4391582 DOI: 10.1186/s13195-015-0105-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 02/06/2015] [Indexed: 11/29/2022]
Abstract
Introduction Alzheimer’s disease (AD) is a growing socioeconomic problem worldwide. Early diagnosis and prevention of this devastating disease have become a research priority. Consequently, the identification of clinically significant and sensitive blood biomarkers for its early detection is very important. Apolipoprotein E (APOE) is a well-known and established genetic risk factor for late-onset AD; however, the impact of the protein level on AD risk is unclear. We assessed the utility of plasma ApoE protein as a potential biomarker of AD in the large, well-characterised Australian Imaging, Biomarkers and Lifestyle Study of Ageing (AIBL) cohort. Methods Total plasma ApoE levels were measured at 18-month follow-up using a commercial bead-based enzyme-linked immunosorbent assay: the Luminex xMAP human apolipoprotein kit. ApoE levels were then analysed between clinical classifications (healthy controls, mild cognitive impairment (MCI) and AD) and correlated with the data available from the AIBL cohort, including but not limited to APOE genotype and cerebral amyloid burden. Results A significant decrease in ApoE levels was found in the AD group compared with the healthy controls. These results validate previously published ApoE protein levels at baseline obtained using different methodology. ApoE protein levels were also significantly affected, depending on APOE genotypes, with ε2/ε2 having the highest protein levels and ε4/ε4 having the lowest. Plasma ApoE levels were significantly negatively correlated with cerebral amyloid burden as measured by neuroimaging. Conclusions ApoE is decreased in individuals with AD compared with healthy controls at 18-month follow-up, and this trend is consistent with our results published at baseline. The influence of APOE genotype and sex on the protein levels are also explored. It is clear that ApoE is a strong player in the aetiology of this disease at both the protein and genetic levels.
Collapse
Affiliation(s)
- Veer B Gupta
- Centre of Excellence in Alzheimer's Disease Research and Care, School of Medical Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup, 6027 Australia ; McCusker Alzheimer's Research Foundation, Hollywood Medical Centre, 85 Monash Avenue, Suite 22, Nedlands, 6009 Australia
| | - Andrea C Wilson
- Centre of Excellence in Alzheimer's Disease Research and Care, School of Medical Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup, 6027 Australia ; McCusker Alzheimer's Research Foundation, Hollywood Medical Centre, 85 Monash Avenue, Suite 22, Nedlands, 6009 Australia
| | - Samantha Burnham
- CSIRO Computational Informatics, Preventative Health Flagship, 65 Brockway Road, Floreat, 6014 Australia
| | - Eugene Hone
- Centre of Excellence in Alzheimer's Disease Research and Care, School of Medical Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup, 6027 Australia ; McCusker Alzheimer's Research Foundation, Hollywood Medical Centre, 85 Monash Avenue, Suite 22, Nedlands, 6009 Australia
| | - Steve Pedrini
- Centre of Excellence in Alzheimer's Disease Research and Care, School of Medical Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup, 6027 Australia ; McCusker Alzheimer's Research Foundation, Hollywood Medical Centre, 85 Monash Avenue, Suite 22, Nedlands, 6009 Australia
| | - Simon M Laws
- Centre of Excellence in Alzheimer's Disease Research and Care, School of Medical Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup, 6027 Australia ; McCusker Alzheimer's Research Foundation, Hollywood Medical Centre, 85 Monash Avenue, Suite 22, Nedlands, 6009 Australia ; Cooperative Research Centre for Mental Health, Carlton, VIC 3053 Australia
| | - Wei Ling Florence Lim
- Centre of Excellence in Alzheimer's Disease Research and Care, School of Medical Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup, 6027 Australia ; McCusker Alzheimer's Research Foundation, Hollywood Medical Centre, 85 Monash Avenue, Suite 22, Nedlands, 6009 Australia
| | - Alan Rembach
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC 3052 Australia
| | - Stephanie Rainey-Smith
- Centre of Excellence in Alzheimer's Disease Research and Care, School of Medical Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup, 6027 Australia ; McCusker Alzheimer's Research Foundation, Hollywood Medical Centre, 85 Monash Avenue, Suite 22, Nedlands, 6009 Australia
| | - David Ames
- Academic Unit for Psychiatry of Old Age, Department of Psychiatry, The University of Melbourne, St Vincent's Aged Psychiatry Service, St George's Hospital, Melbourne, VIC 3065 Australia ; National Ageing Research Institute, Parkville, VIC 3052 Australia
| | - Lynne Cobiac
- CSIRO Preventative Health Flagship, Adelaide, SA 5000 Australia
| | | | - Colin L Masters
- Cooperative Research Centre for Mental Health, Carlton, VIC 3053 Australia ; Mental Health Research Institute, The University of Melbourne, Parkville, VIC 3052 Australia ; Centre for Neuroscience, The University of Melbourne, Parkville, VIC 3010 Australia
| | - Christopher C Rowe
- Department of Nuclear Medicine & Centre for PET, Austin Health, Heidelberg, VIC 3084 Australia
| | - Ashley I Bush
- Cooperative Research Centre for Mental Health, Carlton, VIC 3053 Australia ; Mental Health Research Institute, The University of Melbourne, Parkville, VIC 3052 Australia ; Centre for Neuroscience, The University of Melbourne, Parkville, VIC 3010 Australia
| | - Ralph N Martins
- Centre of Excellence in Alzheimer's Disease Research and Care, School of Medical Sciences, Edith Cowan University, 270 Joondalup Drive, Joondalup, 6027 Australia ; McCusker Alzheimer's Research Foundation, Hollywood Medical Centre, 85 Monash Avenue, Suite 22, Nedlands, 6009 Australia ; Cooperative Research Centre for Mental Health, Carlton, VIC 3053 Australia ; School of Psychiatry and Clinical Neurosciences, The University of Western Australia, Nedlands, 6009 Australia
| | | |
Collapse
|
47
|
Raj A, LoCastro E, Kuceyeski A, Tosun D, Relkin N, Weiner M. Network Diffusion Model of Progression Predicts Longitudinal Patterns of Atrophy and Metabolism in Alzheimer's Disease. Cell Rep 2015; 10:359-369. [PMID: 25600871 DOI: 10.1016/j.celrep.2014.12.034] [Citation(s) in RCA: 126] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 11/08/2014] [Accepted: 12/15/2014] [Indexed: 01/18/2023] Open
Abstract
Alzheimer's disease pathology (AD) originates in the hippocampus and subsequently spreads to temporal, parietal, and prefrontal association cortices in a relatively stereotyped progression. Current evidence attributes this orderly progression to transneuronal transmission of misfolded proteins along the projection pathways of affected neurons. A network diffusion model was recently proposed to mathematically predict disease topography resulting from transneuronal transmission on the brain's connectivity network. Here, we use this model to predict future patterns of regional atrophy and metabolism from baseline regional patterns of 418 subjects. The model accurately predicts end-of-study regional atrophy and metabolism starting from baseline data, with significantly higher correlation strength than given by the baseline statistics directly. The model's rate parameter encapsulates overall atrophy progression rate; group analysis revealed this rate to depend on diagnosis as well as baseline cerebrospinal fluid (CSF) biomarker levels. This work helps validate the model as a prognostic tool for Alzheimer's disease assessment.
Collapse
Affiliation(s)
- Ashish Raj
- Department of Radiology, Weill Medical College of Cornell University, 515 East 71 Street, Suite S123, New York, NY 10021, USA.
| | - Eve LoCastro
- Department of Radiology, Weill Medical College of Cornell University, 515 East 71 Street, Suite S123, New York, NY 10021, USA
| | - Amy Kuceyeski
- Department of Radiology, Weill Medical College of Cornell University, 515 East 71 Street, Suite S123, New York, NY 10021, USA
| | - Duygu Tosun
- Department of Radiology and Biomedical Imaging, Center for Imaging of Neurodegenerative Diseases, University of California, San Francisco, 4150 Clement Street (114M), San Francisco, CA 94121, USA
| | - Norman Relkin
- Department of Neurology and Neuroscience, Memory Disorders Program, Weill Medical College of Cornell University, 428 East 72nd Street, Suite 500, New York, NY 10021, USA
| | - Michael Weiner
- Department of Radiology and Biomedical Imaging, Center for Imaging of Neurodegenerative Diseases, University of California, San Francisco, 4150 Clement Street (114M), San Francisco, CA 94121, USA
| |
Collapse
|
48
|
DeCarlo CA, Tuokko HA, Williams D, Dixon RA, MacDonald SWS. BioAge: toward a multi-determined, mechanistic account of cognitive aging. Ageing Res Rev 2014; 18:95-105. [PMID: 25278166 PMCID: PMC4258131 DOI: 10.1016/j.arr.2014.09.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 09/06/2014] [Accepted: 09/15/2014] [Indexed: 01/15/2023]
Abstract
The search for reliable early indicators of age-related cognitive decline represents a critical avenue for progress in aging research. Chronological age is a commonly used developmental index; however, it offers little insight into the mechanisms underlying cognitive decline. In contrast, biological age (BioAge), reflecting the vitality of essential biological systems, represents a promising operationalization of developmental time. Current BioAge models have successfully predicted age-related cognitive deficits. Research on aging-related cognitive function indicates that the interaction of multiple risk and protective factors across the human lifespan confers individual risk for late-life cognitive decline, implicating a multi-causal explanation. In this review, we explore current BioAge models, describe three broad yet pathologically relevant biological processes linked to cognitive decline, and propose a novel operationalization of BioAge accounting for both moderating and causal mechanisms of cognitive decline and dementia. We argue that a multivariate and mechanistic BioAge approach will lead to a greater understanding of disease pathology as well as more accurate prediction and early identification of late-life cognitive decline.
Collapse
Affiliation(s)
- Correne A DeCarlo
- Department of Psychology, University of Victoria, Victoria, BC, Canada; Centre on Aging, University of Victoria, Victoria, BC, Canada.
| | - Holly A Tuokko
- Department of Psychology, University of Victoria, Victoria, BC, Canada; Centre on Aging, University of Victoria, Victoria, BC, Canada
| | - Dorothy Williams
- Department of Geriatrics, West Coast General Hospital, Port Alberni, BC, Canada
| | - Roger A Dixon
- Department of Psychology, University of Alberta, Edmonton, AB Canada
| | - Stuart W S MacDonald
- Department of Psychology, University of Victoria, Victoria, BC, Canada; Centre on Aging, University of Victoria, Victoria, BC, Canada
| |
Collapse
|
49
|
Tai LM, Koster KP, Luo J, Lee SH, Wang YT, Collins NC, Ben Aissa M, Thatcher GRJ, LaDu MJ. Amyloid-β pathology and APOE genotype modulate retinoid X receptor agonist activity in vivo. J Biol Chem 2014; 289:30538-30555. [PMID: 25217640 DOI: 10.1074/jbc.m114.600833] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Previous data demonstrate that bexarotene (Bex), retinoid X receptor (RXR) agonist, reduces soluble and insoluble amyloid-β (Aβ) in Alzheimer disease (AD)-transgenic mice either by increasing the levels of mouse apolipoprotein E (apoE) or increasing ABCA1/ABCG1-induced apoE lipoprotein association/lipidation. However, although the mechanism of action of RXR agonists remains unclear, a major concern for their use is human (h)-APOE4, the greatest AD genetic risk factor. If APOE4 imparts a toxic gain-of-function, then increasing apoE4 may increase soluble Aβ, likely the proximal AD neurotoxin. If the APOE4 loss-of-function is lipidation of apoE4, then induction of ABCA1/ABCG1 may be beneficial. In novel EFAD-Tg mice (overexpressing h-Aβ42 with h-APOE), levels of soluble Aβ (Aβ42 and oligomeric Aβ) are highest in E4FAD hippocampus (HP) > E3FAD-HP > E4FAD cortex (CX) > E3FAD-CX, whereas levels of lipoprotein-associated/lipidated apoE have the opposite pattern (6 months). In E4FAD-HP, short-term RXR agonist treatment (Bex or LG100268; 5.75-6 months) increased ABCA1, apoE4 lipoprotein-association/lipidation, and apoE4/Aβ complex, decreased soluble Aβ, and increased PSD95. In addition, hydrogel delivery, which mimics low sustained release, was equally effective as gavage for Bex and LG100268. RXR agonists induced no beneficial effects in the E4FAD-HP in a prevention protocol (5-6 months) and actually increased soluble Aβ levels in E3FAD-CX and E4FAD-CX with the short-term protocol, possibly the result of systemic hepatomegaly. Thus, RXR agonists address the loss-of-function associated with APOE4 and exacerbated by Aβ pathology, i.e. low levels of apoE4 lipoprotein association/lipidation. Further studies are vital to address whether RXR agonists are an APOE4-specific AD therapeutic and the systemic side effects that limit translational application.
Collapse
Affiliation(s)
- Leon M Tai
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, Illinois 60612
| | - Kevin P Koster
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, Illinois 60612
| | - Jia Luo
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, and University of Illinois at Chicago, Chicago, Illinois 60612
| | - Sue H Lee
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, and University of Illinois at Chicago, Chicago, Illinois 60612
| | - Yue-Ting Wang
- UICentre (Drug Discovery at UIC), University of Illinois at Chicago, Chicago, Illinois 60612
| | - Nicole C Collins
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, Illinois 60612
| | - Manel Ben Aissa
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, Illinois 60612
| | - Gregory R J Thatcher
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, and University of Illinois at Chicago, Chicago, Illinois 60612
| | - Mary Jo LaDu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, Illinois 60612.
| |
Collapse
|
50
|
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
|