1
|
Ng TKS, Beck T, Liu X, Desai P, Holland T, Dhana K, Krueger K, Wilson RS, Evans DA, Rajan KB. Longitudinal associations between lipid panel and cognitive decline modified by APOE 4 carrier status in biracial community-dwelling older adults: Findings from the Chicago health and aging project. Arch Gerontol Geriatr 2025; 134:105825. [PMID: 40179541 PMCID: PMC12058384 DOI: 10.1016/j.archger.2025.105825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 02/25/2025] [Accepted: 03/08/2025] [Indexed: 04/05/2025]
Abstract
BACKGROUND There have been contradictory findings on the associations between lipids and cognitive decline (CD), which may be attributed to the heterogeneity in the APOE4 carrier status, given APOE's lipid transportation roles. However, extant studies rarely examined the modifying effects of APOE4 carrier status on the associations between lipids and CD. METHODS We analyzed the Chicago Health and Aging Project, a 20-year cohort study comprising older adults with lipid panel assayed, i.e., total cholesterol (TC), triglycerides (TG), high-density lipoprotein (HDL), and low-density lipoprotein (LDL), and longitudinal cognitive tests. We ran adjusted linear mixed-effects models, regressing cognitive test composite on each of the four lipids independently, first with the total sample and subsequently using interaction and stratified subgroup analyses, examining the modifying effects of APOE4 carrier status on the associations. RESULTS 3,496 biracial community-dwelling older adults were recruited from the South side of Chicago (58% African American & 64% women; mean follow-up = 4.6 years). In the total sample, there was a borderline association between TG and CD, estimate (SD, p-value) = 0.0001 (0.0000,0.0565). No associations were detected with other lipids. In the interaction and subgroup analyses, only in ε4 carriers that higher TC levels were significantly associated with accelerated CD, -0.020 (0.009,0.035), whereas higher TG levels were significantly associated with decelerated CD, 0.001 (0.001,0.045). No modifying effects of ε4 carrier status were detected with other lipids. DISCUSSION Specific lipids, i.e., TC and TG, were associated with CD only in the ε4 carriers, highlighting the potential importance of measuring APOE4 status to better inform risk prediction and treatment.
Collapse
Affiliation(s)
- Ted K S Ng
- Rush Institute for Healthy Aging, Department of Internal Medicine, Rush University Medical Center, Chicago, IL, USA.
| | - Todd Beck
- Rush Institute for Healthy Aging, Department of Internal Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Xiaoran Liu
- Rush Institute for Healthy Aging, Department of Internal Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Pankaja Desai
- Rush Institute for Healthy Aging, Department of Internal Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Thomas Holland
- Rush University Medical Center, Rush Institute for Healthy Aging & College of Health Sciences, USA
| | - Klodian Dhana
- Rush Institute for Healthy Aging, Department of Internal Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Kristin Krueger
- Rush Institute for Healthy Aging, Department of Internal Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Robert S Wilson
- Rush Alzheimer's Disease Research Center, Department of Internal Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Denis A Evans
- Rush Institute for Healthy Aging, Department of Internal Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Kumar B Rajan
- Rush Institute for Healthy Aging, Department of Internal Medicine, Rush University Medical Center, Chicago, IL, USA; Rush Alzheimer's Disease Research Center, Department of Internal Medicine, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
2
|
Inan S, Wilson RP, Tükel Ç. IUPHAR review: From gut to brain: The role of gut dysbiosis, bacterial amyloids, and metabolic disease in Alzheimer's disease. Pharmacol Res 2025; 215:107693. [PMID: 40086611 DOI: 10.1016/j.phrs.2025.107693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/06/2025] [Accepted: 03/07/2025] [Indexed: 03/16/2025]
Abstract
Gut microbial dysbiosis, or altered gut microbial communities, in Alzheimer's Disease suggests a pathogenic role for gut inflammation and microbial products in shaping a neuroinflammatory environment. Similarly, metabolic diseases, such as obesity and diabetes, are also associated with an increased risk of Alzheimer's Disease. As the metabolic landscape shifts during gut inflammation, and gut inflammation in turn impacts metabolic processes, we explore how these interconnected pathways may contribute to the progression of Alzheimer's Disease. Additionally, we discuss the role of bacterial amyloids produced by gut microbes, which may exacerbate amyloid aggregation in the brain and contribute to neurodegenerative processes. Furthermore, we highlight potential therapeutic strategies aimed at reducing gut inflammation, improving metabolic health, and decreasing amyloid content as a means to mitigate Alzheimer's Disease progression. These approaches, targeting the gut-brain-metabolic axis, could offer promising avenues for delaying or preventing cognitive decline in affected individuals.
Collapse
Affiliation(s)
- Saadet Inan
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.
| | - R Paul Wilson
- Center for Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Çagla Tükel
- Center for Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
3
|
Ray NR, Bradley J, Yilmaz E, Kizil C, Kurup JT, Martin ER, Klein HU, Kunkle BW, Bennett DA, De Jager PL, Alzheimer’s Disease Genetics Consortium, Beecham GW, Cruchaga C, Reitz C. Local genetic covariance analysis with lipid traits identifies novel loci for early-onset Alzheimer's Disease. PLoS Genet 2025; 21:e1011631. [PMID: 40096060 PMCID: PMC11984970 DOI: 10.1371/journal.pgen.1011631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 04/10/2025] [Accepted: 02/19/2025] [Indexed: 03/19/2025] Open
Abstract
The genetic component of early-onset Alzheimer disease (EOAD), accounting for ~10% of all Alzheimer's disease (AD) cases, is largely unexplained. Recent studies suggest that EOAD may be enriched for variants acting in the lipid pathway. The current study examines the shared genetic heritability between EOAD and the lipid pathway using genome-wide multi-trait genetic covariance analyses. Summary statistics were obtained from the GWAS meta-analyses of EOAD by the Alzheimer's Disease Genetics Consortium (n=19,668) and five blood lipid traits by the Global Lipids Genetics Consortium (n=1,320,016). The significant results were compared between the EOAD and lipids GWAS and genetic covariance analyses were performed via SUPERGNOVA. Genes in linkage disequilibrium (LD) with top EOAD hits in identified regions of covariance with lipid traits were scored and ranked for causality by combining evidence from gene-based analysis, AD-risk scores incorporating transcriptomic and proteomic evidence, eQTL data, eQTL colocalization analyses, DNA methylation data, and single-cell RNA sequencing analyses. Direct comparison of GWAS results showed 5 loci overlapping between EOAD and at least one lipid trait harboring APOE, TREM2, MS4A4E, LILRA5, and LRRC25. Local genetic covariance analyses identified 3 regions of covariance between EOAD and at least one lipid trait. Gene prioritization nominated 3 likely causative genes at these loci: ANKDD1B, CUZD1, and MS4A64.The current study identified genetic covariance between EOAD and lipids, providing further evidence of shared genetic architecture and mechanistic pathways between the two traits.
Collapse
Affiliation(s)
- Nicholas R. Ray
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, New York, United States of America
- Gertrude H. Sergievsky Center, Columbia University, New York, New York, United States of America
| | - Joseph Bradley
- NeuroGenomics and Informatics Center, Washington University School of Medicine in St. Louis, St. Louis, Missouri, United States of America
| | - Elanur Yilmaz
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, New York, United States of America
- Gertrude H. Sergievsky Center, Columbia University, New York, New York, United States of America
| | - Caghan Kizil
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, New York, United States of America
- Gertrude H. Sergievsky Center, Columbia University, New York, New York, United States of America
| | - Jiji T. Kurup
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, New York, United States of America
- Gertrude H. Sergievsky Center, Columbia University, New York, New York, United States of America
| | - Eden R. Martin
- The John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida, United States of America
| | - Hans-Ulrich Klein
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, New York, United States of America
- Gertrude H. Sergievsky Center, Columbia University, New York, New York, United States of America
| | - Brian W. Kunkle
- The John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida, United States of America
| | - David A. Bennett
- RUSH Alzheimer’s Disease Center, RUSH University, Chicago, Illinois, United States of America
| | - Philip L. De Jager
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, New York, United States of America
- Gertrude H. Sergievsky Center, Columbia University, New York, New York, United States of America
| | | | - Gary W. Beecham
- The John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida, United States of America
| | - Carlos Cruchaga
- NeuroGenomics and Informatics Center, Washington University School of Medicine in St. Louis, St. Louis, Missouri, United States of America
| | - Christiane Reitz
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, New York, United States of America
- Gertrude H. Sergievsky Center, Columbia University, New York, New York, United States of America
| |
Collapse
|
4
|
Rentschler KM, Kodavanti UP. Mechanistic insights regarding neuropsychiatric and neuropathologic impacts of air pollution. Crit Rev Toxicol 2024; 54:953-980. [PMID: 39655487 PMCID: PMC12043015 DOI: 10.1080/10408444.2024.2420972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 10/07/2024] [Accepted: 10/15/2024] [Indexed: 12/24/2024]
Abstract
Air pollution is a significant environmental health risk for urban areas and developing countries. Air pollution may contribute to the incidence of cardiopulmonary and metabolic diseases. Evidence also points to the role of air pollution in worsening or developing neurological and neuropsychiatric conditions. Inhaled pollutants include compositionally differing mixtures of respirable gaseous and particulate components of varied sizes, solubilities, and chemistry. Inhalation of combustibles and volatile organic compounds (VOCs) or other irritant particulate matter (PM) may trigger lung sensory afferents which initiate a sympathetic stress response via activation of the hypothalamic-pituitary-adrenal (HPA) and sympathetic-adrenal-medullary (SAM) axes. Activation of SAM and HPA axes are associated with selective inhibition of hypothalamic-pituitary-gonadal (HPG) and hypothalamic-pituitary-thyroid (HPT) axes following exposure. Regarding chronic exposure in susceptible hosts, these changes may become pathological by causing neuroinflammation, neurotransmitter, and neuroendocrine imbalances. Soluble PM, such as metals and nano-size particles may translocate across the olfactory, trigeminal, or vagal nerves through retrograde axonal transport, or through systemic circulation which may disrupt the blood-brain barrier (BBB) and deposit in neural tissue. Neuronal deposition of metallic components can have a negative impact through multiple molecular mechanisms. In addition to systemic translocation, the release of pituitary and stress hormones, altered metabolic hormonal status and resultant circulating metabolic milieu, and sympathetically and HPA-mediated changes in immune markers, may secondarily impact the brain through a variety of regulatory adrenal hormone-dependent mechanisms. Several reviews covering air pollution as a risk factor for neuropsychiatric disorders have been published, but no reviews discuss the in-depth intersection between molecular and stress-related neuroendocrine mechanisms, thereby addressing adaptation and susceptibility variations and link to peripheral tissue effects. The purpose of this review is to discuss evidence regarding neurochemical, neuroendocrine, and molecular mechanisms which may contribute to neuropathology from air pollution exposure. This review also covers bi-directional neural and systemic interactions which may raise the risk for air pollution-related systemic illness.
Collapse
Affiliation(s)
- Katherine M. Rentschler
- Oak Ridge Institute for Science and Education Research Participation Program, U.S. Environmental Protection Agency, Research Triangle Park, NC, United States of America
| | - Urmila P. Kodavanti
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina, United States of America
| |
Collapse
|
5
|
Huang LC, Lee MY, Chien CF, Chang YP, Li KY, Yang YH. Age and sex differences in the association between APOE genotype and Alzheimer's disease in a Taiwan Chinese population. Front Aging Neurosci 2023; 15:1246592. [PMID: 37680541 PMCID: PMC10481952 DOI: 10.3389/fnagi.2023.1246592] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 08/07/2023] [Indexed: 09/09/2023] Open
Abstract
Introduction The Apolipoprotein E (APOE) epsilon (ε) 4 allele is a well-established risk factor for late-onset Alzheimer's disease (AD). Reports on white ancestry populations have showed that age, sex, and ethnicity have different effects on the association between APOE genotype and AD. However, studies on Asian populations such as Taiwan Chinese populations are limited. This study aimed to evaluate the association between APOE genotype and AD in a Taiwan Chinese population, and to explore if the association varies by age and sex. Methods We conducted a case-control study in 725 patients with AD and 1,067 age- and sex- matched controls without dementia from a Taiwan Chinese population. Logistic regression models were used to test the association between AD and APOE genotypes. Secondary analyses considered age (<75 or ≥75 years old), and sex stratified models. Results The risk of AD was significantly increased for people with at least one copy of APOE ε4 (OR = 2.52, 95% CI = 2.01-3.17, p < 0.001) and in a dose-dependent manner. Our results did not show an statistically significance different in AD risk when women and men carrying APOEε4 were compared. Despite not reaching statistical significance, the risk of APOE ε4 for AD was higher among younger participants (OR = 3.21, 95% CI = 2.26-4.56, p < 0.001) compared to older ones (OR = 2.13, 95% CI = 1.53-2.97, p < 0.001). When considering both sex and age, the risk of AD was higher among older men carrying APOE ε4 (OR = 2.64, 95% CI = 1.51-4.60 in men; OR = 1.90, 95% CI = 1.26-2.86 in women), while women carrying APOE ε4 appeared to have an increased risk at a younger age (OR = 3.29, 95% CI = 2.20-4.93 in women; OR = 2.91, 95% CI = 1.40-6.05 in men). Discussion The APOE ε4 allele represents a major risk factor for AD in the Taiwanese population. The effect of APOE ε4 allele on AD risk appeared to be stronger among men aged 75 years or more and among younger women.
Collapse
Affiliation(s)
- Ling-Chun Huang
- Department of Neurology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Neuroscience Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Mei-Yueh Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ching-Fang Chien
- Department of Neurology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Neuroscience Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yang-Pei Chang
- Department of Neurology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Neuroscience Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Kuan-Ying Li
- Department of Neurology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Neuroscience Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yuan-Han Yang
- Department of Neurology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Neuroscience Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- School of Post-Baccalaureate Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
6
|
Hjazi A, Ahsan M, Alghamdi MI, Kareem AK, Al-Saidi DN, Qasim MT, Romero-Parra RM, Zabibah RS, Ramírez-Coronel AA, Mustafa YF, Hosseini-Fard SR, Karampoor S, Mirzaei R. Unraveling the impact of 27-hydroxycholesterol in autoimmune diseases: Exploring promising therapeutic approaches. Pathol Res Pract 2023; 248:154737. [PMID: 37542860 DOI: 10.1016/j.prp.2023.154737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 08/01/2023] [Accepted: 08/02/2023] [Indexed: 08/07/2023]
Abstract
The role of 27-hydroxycholesterol (27-OHC) in autoimmune diseases has become a subject of intense research in recent years. This oxysterol, derived from cholesterol, has been identified as a significant player in modulating immune responses and inflammation. Its involvement in autoimmune pathogenesis has drawn attention to its potential as a therapeutic target for managing autoimmune disorders effectively. 27-OHC, an oxysterol derived from cholesterol, has emerged as a key player in modulating immune responses and inflammatory processes. It exerts its effects through various mechanisms, including activation of nuclear receptors, interaction with immune cells, and modulation of neuroinflammation. Additionally, 27-OHC has been implicated in the dysregulation of lipid metabolism, neurotoxicity, and blood-brain barrier (BBB) disruption. Understanding the intricate interplay between 27-OHC and autoimmune diseases, particularly neurodegenerative disorders, holds promise for developing targeted therapeutic strategies. Additionally, emerging evidence suggests that 27-OHC may interact with specific receptors and transcription factors, thus influencing gene expression and cellular processes in autoimmune disorders. Understanding the intricate mechanisms by which 27-OHC influences immune dysregulation and tissue damage in autoimmune diseases is crucial for developing targeted therapeutic interventions. Further investigations into the molecular pathways and signaling networks involving 27-OHC are warranted to unravel its full potential as a therapeutic target in autoimmune diseases, thereby offering new avenues for disease intervention and management.
Collapse
Affiliation(s)
- Ahmed Hjazi
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Maria Ahsan
- King Edward Medical University Lahore, Pakistan
| | - Mohammed I Alghamdi
- Department of Computer Science, Al-Baha University, Al-Baha City, Kingdom of Saudi Arabia
| | - A K Kareem
- Biomedical Engineering Department, Al-Mustaqbal University College, Babylon, Iraq
| | - Dahlia N Al-Saidi
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq
| | - Maytham T Qasim
- Department of Anesthesia, College of Health and Medical Technology, Al-Ayen University, Thi-Qar, Iraq
| | | | - Rahman S Zabibah
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | - Andrés Alexis Ramírez-Coronel
- Health and Behavior Research Group (HBR), Psychometry and Ethology Laboratory, Catholic University of Cuenca, Ecuador; University of Palermo, Buenos Aires, Argentina; Research group in educational statistics, National University of Education, Azogues, Ecuador; Epidemiology and Biostatistics Research Group, CES University, Colombia
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul 41001, Iraq
| | - Seyed Reza Hosseini-Fard
- Department of Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Sajad Karampoor
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Rasoul Mirzaei
- Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
7
|
Wingo AP, Vattathil SM, Liu J, Fan W, Cutler DJ, Levey AI, Schneider JA, Bennett DA, Wingo TS. LDL cholesterol is associated with higher AD neuropathology burden independent of APOE. J Neurol Neurosurg Psychiatry 2022; 93:jnnp-2021-328164. [PMID: 35772923 PMCID: PMC9380478 DOI: 10.1136/jnnp-2021-328164] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 06/07/2022] [Indexed: 11/29/2022]
Abstract
OBJECTIVE APOE is a strong risk factor for Alzheimer's disease (AD) and associated with higher low-density lipoprotein cholesterol (LDL-C) levels. Moreover, LDL-C is associated with the development of clinically ascertained AD; however, whether this association is present with the underlying neuropathological manifestations of AD or whether it is independent of the effect of APOE is unknown and is the focus of this paper. METHODS Individuals in the Religious Orders Study/Memory and Ageing Project cohorts with longitudinal measures of blood lipids and detailed autopsies were studied. We modelled the relationship between blood lipids and 12 age-related brain pathologies using a linear mixed model adjusted for potential confounding factors and stratified by APOE genotype with overall significance determined by meta-analysis. Blood lipids considered were LDL-C, high-density lipoprotein cholesterol and triglycerides. Brain pathologies included AD pathology measured by silver staining (Braak stage, a modified Consortium to Establish a Registry for Alzheimer's Disease [CERAD] score and global AD pathology) and immunohistochemistry (beta-amyloid and neurofibrillary tangles) as well as cerebral microinfarct, cerebral macroinfarct, cerebral amyloid angiopathy, cerebral atherosclerosis, hippocampal sclerosis, TDP-43 cytoplasmic inclusions and Lewy bodies. RESULTS 559 participants (69.1% female) had complete data for analysis. They were followed for a median of 7 years and a median of 3 years prior to dementia onset. LDL-C was associated with all measures of AD neuropathology (neurofibrillary tangles, beta-amyloid, Braak stage, modified CERAD score and global AD pathology) and cerebral amyloid angiopathy independent of APOE after adjusting for age, sex, cholesterol-lowering medication use, body mass index, smoking and education at false discovery rate (FDR) p-value <0.05. CONCLUSIONS These findings implicate LDL-C in the pathophysiology of AD independent of APOE and suggest LDL-C is a modifiable risk factor for AD.
Collapse
Affiliation(s)
- Aliza P Wingo
- Division of Mental Health, Atlanta VA Medical Center, Decatur, Georgia, USA
- Department of Psychiatry, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Selina M Vattathil
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Jiaqi Liu
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Wen Fan
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - David J Cutler
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Allan I Levey
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, USA
- Goizueta Alzheimer's Disease Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Julie A Schneider
- Departments of Pathology and Neurological Science, Rush Alzheimer's Disease Center, Rush University, Chicago, Illinois, USA
- Rush Alzheimer's Disease Center, Rush University, Chicago, Illinois, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University, Chicago, Illinois, USA
| | - Thomas S Wingo
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, USA
- Goizueta Alzheimer's Disease Center, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
8
|
Wang S, Wang X, Wang Z, Wang Z, Jiang L, Liu J, Wu J, Liu Y. Highly sensitive and selective detection of cytochrome P450 46A1 activity by a ultra-high-performance liquid chromatography-tandem mass spectrometry method. Biomed Chromatogr 2021; 36:e5291. [PMID: 34854105 DOI: 10.1002/bmc.5291] [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: 09/08/2021] [Revised: 11/11/2021] [Accepted: 11/15/2021] [Indexed: 11/09/2022]
Abstract
Cytochrome P450 46A1 (CYP46A1) is a key enzyme responsible for metabolizing cholesterol to 24-hydroxycholesterol in the brain, and thus might serve as a therapeutic target for several neurodegenerative disorders including Parkinson's disease, Alzheimer's disease and Huntington's disease. However, an applicable, sensitive and reliable method for the precise measurement of CYP46A1 activities in complex biological samples remains limited. In this study, a novel ultra-high-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) method for highly sensitive and selective determination of 24-hydroxycholesterol was developed to characterize CYP46A1 activity. The mass spectrometric detection was performed using multiple reaction monitoring for 24-hydroxcholesterol at m/z 385.2 → 367.2. The limit of quantification for 24-hydroxycholesterol using this UPLC-MS/MS method was as low as 10 nM, which is lower than those reported previously. The method also showed favorable accuracy and precision. Meanwhile, the short- and long-term stability of this method was fully validated. In addition, the method was successfully applied to investigate the kinetic properties of 24-hydroxycholesterol formation by CYP46A1.
Collapse
Affiliation(s)
- Shujuan Wang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Xiaoyu Wang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Zhe Wang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Zhen Wang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Lili Jiang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Jing Liu
- College of Pharmacy, Dalian Medical University, Dalian, China
| | - Jingjing Wu
- College of Pharmacy, Dalian Medical University, Dalian, China
| | - Yong Liu
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| |
Collapse
|
9
|
Wang ZH, Xia Y, Liu P, Liu X, Edgington-Mitchell L, Lei K, Yu SP, Wang XC, Ye K. ApoE4 activates C/EBPβ/δ-secretase with 27-hydroxycholesterol, driving the pathogenesis of Alzheimer's disease. Prog Neurobiol 2021; 202:102032. [PMID: 33716161 DOI: 10.1016/j.pneurobio.2021.102032] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 01/17/2021] [Accepted: 03/09/2021] [Indexed: 02/06/2023]
Abstract
ApoE4, an apolipoprotein implicated in cholesterol transport and amyloid-β (Aβ) metabolism, is a major genetic risk determinant for Alzheimer's Disease (AD) and drives its pathogenesis via Aβ-dependent and -independent pathways. C/EBPβ, a proinflammatory cytokines-activated transcription factor, is upregulated in AD and mediates cytokines and δ-secretase expression. However, how ApoE4 contributes to AD pathogenesis remains incompletely understood. Here we show that ApoE4 and 27-hydroxycholesterol (27-OHC) co-activate C/EBPβ/δ-secretase signaling in neurons, mediating AD pathogenesis, and this effect is dependent on neuronal secreted Aβ and inflammatory cytokines. Inhibition of cholesterol metabolism with lovastatin diminishes neuronal ApoE4's stimulatory effects. Furthermore, ApoE4 and 27-OHC also mediate lysosomal δ-secretase leakage, activation, secretion and endocytosis. Notably, 27-OHC strongly activates C/EBPβ/δ-secretase pathway in human ApoE4-TR mice and triggers AD pathologies and cognitive deficits, which is blocked by C/EBPβ depletion. Hence, our findings demonstrate that ApoE4 and 27-OHC additively trigger AD pathogenesis via activating C/EBPβ/δ-secretase pathway. Lowering cholesterol levels with statins should benefit the ApoE4 AD carriers.
Collapse
Affiliation(s)
- Zhi-Hao Wang
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, 30322, USA; Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yiyuan Xia
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, 30322, USA; Department of Pathophysiology, Key Laboratory of Ministry of Education of Neurological Diseases, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pai Liu
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, 30322, USA; Neuroscience Graduate Program, Laney Graduate School, Emory University, Atlanta, GA, 30322, USA
| | - Xia Liu
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Laura Edgington-Mitchell
- Department of Biochemistry and Molecular Biology, The University of Melbourne, Melbourne Victoria, 3010, Australia
| | - Kecheng Lei
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Shan Ping Yu
- Department of Anesthesiology, Emory University School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Xiao-Chuan Wang
- Department of Pathophysiology, Key Laboratory of Ministry of Education of Neurological Diseases, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, JS, 226001, China.
| | - Keqiang Ye
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
10
|
Nicolosi M, Bellia F, Giuffrida ML, Zimbone S, Oliveri V, Vecchio G. Synthesis and biological evaluation of novel β-cyclodextrin-fluvastatin conjugates. RESULTS IN CHEMISTRY 2021. [DOI: 10.1016/j.rechem.2021.100230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
11
|
Saddiki H, Fayosse A, Cognat E, Sabia S, Engelborghs S, Wallon D, Alexopoulos P, Blennow K, Zetterberg H, Parnetti L, Zerr I, Hermann P, Gabelle A, Boada M, Orellana A, de Rojas I, Lilamand M, Bjerke M, Van Broeckhoven C, Farotti L, Salvadori N, Diehl-Schmid J, Grimmer T, Hourregue C, Dugravot A, Nicolas G, Laplanche JL, Lehmann S, Bouaziz-Amar E, the Alzheimer’s Disease Neuroimaging Initiative, Hugon J, Tzourio C, Singh-Manoux A, Paquet C, Dumurgier J. Age and the association between apolipoprotein E genotype and Alzheimer disease: A cerebrospinal fluid biomarker-based case-control study. PLoS Med 2020; 17:e1003289. [PMID: 32817639 PMCID: PMC7446786 DOI: 10.1371/journal.pmed.1003289] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 07/22/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The ε4 allele of apolipoprotein E (APOE) gene and increasing age are two of the most important known risk factors for developing Alzheimer disease (AD). The diagnosis of AD based on clinical symptoms alone is known to have poor specificity; recently developed diagnostic criteria based on biomarkers that reflect underlying AD neuropathology allow better assessment of the strength of the associations of risk factors with AD. Accordingly, we examined the global and age-specific association between APOE genotype and AD by using the A/T/N classification, relying on the cerebrospinal fluid (CSF) levels of β-amyloid peptide (A, β-amyloid deposition), phosphorylated tau (T, pathologic tau), and total tau (N, neurodegeneration) to identify patients with AD. METHODS AND FINDINGS This case-control study included 1,593 white AD cases (55.4% women; mean age 72.8 [range = 44-96] years) with abnormal values of CSF biomarkers from nine European memory clinics and the American Alzheimer's Disease Neuroimaging Initiative (ADNI) study. A total of 11,723 dementia-free controls (47.1% women; mean age 65.6 [range = 44-94] years) were drawn from two longitudinal cohort studies (Whitehall II and Three-City), in which incident cases of dementia over the follow-up were excluded from the control population. Odds ratio (OR) and population attributable fraction (PAF) for AD associated with APOE genotypes were determined, overall and by 5-year age categories. In total, 63.4% of patients with AD and 22.6% of population controls carried at least one APOE ε4 allele. Compared with non-ε4 carriers, heterozygous ε4 carriers had a 4.6 (95% confidence interval 4.1-5.2; p < 0.001) and ε4/ε4 homozygotes a 25.4 (20.4-31.2; p < 0.001) higher OR of AD in unadjusted analysis. This association was modified by age (p for interaction < 0.001). The PAF associated with carrying at least one ε4 allele was greatest in the 65-70 age group (69.7%) and weaker before 55 years (14.2%) and after 85 years (22.6%). The protective effect of APOE ε2 allele for AD was unaffected by age. Main study limitations are that analyses were based on white individuals and AD cases were drawn from memory centers, which may not be representative of the general population of patients with AD. CONCLUSIONS In this study, we found that AD diagnosis based on biomarkers was associated with APOE ε4 carrier status, with a higher OR than previously reported from studies based on only clinical AD criteria. This association differs according to age, with the strongest effect at 65-70 years. These findings highlight the need for early interventions for dementia prevention to mitigate the effect of APOE ε4 at the population level.
Collapse
Affiliation(s)
- Hana Saddiki
- Université de Paris, Inserm U1153, Epidemiology of Ageing and Neurodegenerative diseases, Paris, France
| | - Aurore Fayosse
- Université de Paris, Inserm U1153, Epidemiology of Ageing and Neurodegenerative diseases, Paris, France
| | - Emmanuel Cognat
- Cognitive Neurology Center, Lariboisiere—Fernand Widal Hospital, AP-HP, Université de Paris, Paris, France
| | - Séverine Sabia
- Université de Paris, Inserm U1153, Epidemiology of Ageing and Neurodegenerative diseases, Paris, France
| | - Sebastiaan Engelborghs
- Department of Neurology, Universitair Ziekenhuis Brussel, Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium
- Department of Biomedical Sciences, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - David Wallon
- Inserm U1245, Rouen University Hospital, Department of Neurology and CNR-MAJ, Normandy Center for Genomic and Personalized Medicine, Rouen, France
| | - Panagiotis Alexopoulos
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, Faculty of Medicine, Technical University of Munich, Munich, Germany
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, Institute of Neurology, University College London, UK Dementia Research Institute, London, United Kingdom
| | - Lucilla Parnetti
- Center for Memory Disturbances-Lab of Clinical Neurochemistry, Section of Neurology, University of Perugia, Italy
| | - Inga Zerr
- Department of Neurology, Clinical Dementia Center, University Medical Center Göttingen and German Center for Neurodegenerative Diseases, Göttingen, Germany
| | - Peter Hermann
- Department of Neurology, Clinical Dementia Center, University Medical Center Göttingen and German Center for Neurodegenerative Diseases, Göttingen, Germany
| | - Audrey Gabelle
- Department of Neurology, Memory Research and Resources Centre, University of Montpellier, Montpellier, France
| | - Mercè Boada
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurciències Aplicades, Universitat International de Catalunya, Barcelona, Spain
| | - Adelina Orellana
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurciències Aplicades, Universitat International de Catalunya, Barcelona, Spain
| | - Itziar de Rojas
- Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurciències Aplicades, Universitat International de Catalunya, Barcelona, Spain
| | - Matthieu Lilamand
- Cognitive Neurology Center, Lariboisiere—Fernand Widal Hospital, AP-HP, Université de Paris, Paris, France
| | - Maria Bjerke
- VIB Center for Molecular Neurology, Institute Born-Bunge and Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Christine Van Broeckhoven
- VIB Center for Molecular Neurology, Institute Born-Bunge and Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Lucia Farotti
- Center for Memory Disturbances-Lab of Clinical Neurochemistry, Section of Neurology, University of Perugia, Italy
| | - Nicola Salvadori
- Center for Memory Disturbances-Lab of Clinical Neurochemistry, Section of Neurology, University of Perugia, Italy
| | - Janine Diehl-Schmid
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, Faculty of Medicine, Technical University of Munich, Munich, Germany
| | - Timo Grimmer
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, Faculty of Medicine, Technical University of Munich, Munich, Germany
| | - Claire Hourregue
- Cognitive Neurology Center, Lariboisiere—Fernand Widal Hospital, AP-HP, Université de Paris, Paris, France
| | - Aline Dugravot
- Université de Paris, Inserm U1153, Epidemiology of Ageing and Neurodegenerative diseases, Paris, France
| | - Gaël Nicolas
- Inserm U1245, Rouen University Hospital, Department of Neurology and CNR-MAJ, Normandy Center for Genomic and Personalized Medicine, Rouen, France
| | - Jean-Louis Laplanche
- Department of Biochemistry and Molecular Biology, Lariboisière Hospital, APHP, Paris, France
| | - Sylvain Lehmann
- Department of Biochemistry, University of Montpellier, Montpellier, France
| | - Elodie Bouaziz-Amar
- Department of Biochemistry and Molecular Biology, Lariboisière Hospital, APHP, Paris, France
| | | | - Jacques Hugon
- Cognitive Neurology Center, Lariboisiere—Fernand Widal Hospital, AP-HP, Université de Paris, Paris, France
| | - Christophe Tzourio
- Bordeaux Population Health Research Center, Team HEALTHY, UMR1219, University of Bordeaux, Inserm, Bordeaux, France
| | - Archana Singh-Manoux
- Université de Paris, Inserm U1153, Epidemiology of Ageing and Neurodegenerative diseases, Paris, France
- Department of Epidemiology and Public Health, University College London, London, United Kingdom
| | - Claire Paquet
- Cognitive Neurology Center, Lariboisiere—Fernand Widal Hospital, AP-HP, Université de Paris, Paris, France
| | - Julien Dumurgier
- Université de Paris, Inserm U1153, Epidemiology of Ageing and Neurodegenerative diseases, Paris, France
- Cognitive Neurology Center, Lariboisiere—Fernand Widal Hospital, AP-HP, Université de Paris, Paris, France
- * E-mail:
| |
Collapse
|
12
|
Wingo TS, Cutler DJ, Wingo AP, Le NA, Rabinovici GD, Miller BL, Lah JJ, Levey AI. Association of Early-Onset Alzheimer Disease With Elevated Low-Density Lipoprotein Cholesterol Levels and Rare Genetic Coding Variants of APOB. JAMA Neurol 2020; 76:809-817. [PMID: 31135820 DOI: 10.1001/jamaneurol.2019.0648] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Importance Early-onset Alzheimer disease (EOAD) is a rare form of Alzheimer disease (AD) with a large genetic basis that is only partially understood. In late-onset AD, elevated circulating cholesterol levels increase AD risk even after adjusting for the apolipoprotein E ε4 (APOE E4) allele, a major genetic factor for AD and elevated cholesterol levels; however, the role of circulating cholesterol levels in EOAD is unclear. Objectives To investigate the association between circulating cholesterol levels and EOAD and to identify genetic variants underlying this possible association. Design, Setting, and Participants In this case series, plasma cholesterol levels were directly measured in 267 samples from the AD research centers (ADRCs) of Emory University and University of California, San Francisco, collected from January 21, 2009, through August 21, 2014. The association between cholesterol and EOAD was examined using multiple linear regression. To determine the underlying genetic variants, APOB, APP, PSEN1, and PSEN2 were sequenced in samples from 2125 EOAD cases and controls recruited from 29 ADRCs from January 1, 1984, through December 31, 2015. Data were analyzed from November 23, 2016, through April 10, 2018. Exposures Clinical diagnosis, age at clinical diagnosis, plasma cholesterol measures (total cholesterol, low-density lipoprotein cholesterol [LDL-C], high-density lipoprotein cholesterol, triglycerides, and apolipoprotein B), and genetic variants in APOE, APP, PSEN1, PSEN2, and APOB. Main Outcomes and Measures The primary outcome was the association between EOAD and plasma cholesterol measures. The secondary outcome was the association between EOAD and the burden of genetic variants in APOB. Results Of the 2125 samples that underwent genetic sequencing, 1276 were from women (60.0%) and 654 (30.8%) were from patients with EOAD (mean [SD] ages, 55.6 [4.3] years for cases and 72.0 [9.6] years for controls). APOE E4 explained 10.1% of the variance of EOAD. After controlling for APOE E4, EOAD cases had higher levels of total cholesterol (mean difference [SE], 21.9 [5.2] mg/dL; P = 2.9 × 10-5), LDL-C (mean difference [SE], 22.0 [4.5] mg/dL; P = 1.8 × 10-6), and ApoB (mean difference [SE], 12.0 [2.4] mg/dL; P = 2.0 × 10-6) than controls in 267 frozen samples. Approximately 3% of EOAD cases carried known AD-causing mutations. Gene-based rare variant burden testing in 2066 samples showed that rare APOB coding variants were significantly more abundant in EOAD cases after adjusting for sex, APOE E4, genetic principal components, ADRC center, and batch (effect size, 0.20; P = 4.20 × 10-4). Conclusions and Relevance Elevated LDL-C levels were associated with higher probability of having EOAD, and EOAD cases were enriched for rare coding variants in APOB, which codes for the major protein of LDL-C. Collectively, these novel findings highlight the important role of LDL-C in EOAD pathogenesis and suggest a direct link of APOB variants to AD risk.
Collapse
Affiliation(s)
- Thomas S Wingo
- Division of Neurology, Atlanta Veterans Affairs Medical Center, Decatur, Georgia.,Department of Neurology, Emory University School of Medicine, Atlanta, Georgia.,Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia
| | - David J Cutler
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia
| | - Aliza P Wingo
- Division of Mental Health, Atlanta Veterans Affairs Medical Center, Decatur, Georgia.,Department of Psychiatry, Emory University School of Medicine, Atlanta, Georgia
| | - Ngoc-Anh Le
- Biomarker Core Laboratory, Atlanta Veterans Affairs Medical Center, Decatur, Georgia
| | - Gil D Rabinovici
- Department of Neurology, University of California, San Francisco
| | - Bruce L Miller
- Department of Neurology, University of California, San Francisco
| | - James J Lah
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia
| | - Allan I Levey
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
13
|
Anderson AE, Diaz-Santos M, Frei S, Dang BH, Kaur P, Lyden P, Buxton R, Douglas PK, Bilder RM, Esfandiari M, Friston KJ, Nookala U, Bookheimer SY. Hemodynamic latency is associated with reduced intelligence across the lifespan: an fMRI DCM study of aging, cerebrovascular integrity, and cognitive ability. Brain Struct Funct 2020; 225:1705-1717. [PMID: 32474754 DOI: 10.1007/s00429-020-02083-w] [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: 10/03/2019] [Accepted: 05/04/2020] [Indexed: 10/24/2022]
Abstract
Changes in neurovascular coupling are associated with both Alzheimer's disease and vascular dementia in later life, but this may be confounded by cerebrovascular risk. We hypothesized that hemodynamic latency would be associated with reduced cognitive functioning across the lifespan, holding constant demographic and cerebrovascular risk. In 387 adults aged 18-85 (mean = 48.82), dynamic causal modeling was used to estimate the hemodynamic response function in the left and right V1 and V3-ventral regions of the visual cortex in response to a simple checkerboard block design stimulus with minimal cognitive demands. The hemodynamic latency (transit time) in the visual cortex was used to predict general cognitive ability (Full-Scale IQ), controlling for demographic variables (age, race, education, socioeconomic status) and cerebrovascular risk factors (hypertension, alcohol use, smoking, high cholesterol, BMI, type 2 diabetes, cardiac disorders). Increased hemodynamic latency in the visual cortex predicted reduced cognitive function (p < 0.05), holding constant demographic and cerebrovascular risk. Increased alcohol use was associated with reduced overall cognitive function (Full Scale IQ 2.8 pts, p < 0.05), while cardiac disorders (Full Scale IQ 3.3 IQ pts; p < 0.05), high cholesterol (Full Scale IQ 3.9 pts; p < 0.05), and years of education (2 IQ pts/year; p < 0.001) were associated with higher general cognitive ability. Increased hemodynamic latency was associated with reduced executive functioning (p < 0.05) as well as reductions in verbal concept formation (p < 0.05) and the ability to synthesize and analyze abstract visual information (p < 0.01). Hemodynamic latency is associated with reduced cognitive ability across the lifespan, independently of other demographic and cerebrovascular risk factors. Vascular health may predict cognitive ability long before the onset of dementias.
Collapse
Affiliation(s)
- Ariana E Anderson
- Department of Psychiatry and Biobehavioral Sciences, University of California, 760 Westwood Plaza, Suite 28-224, Los Angeles, 90095, USA. .,Department of Statistics, University of California, Los Angeles, USA.
| | - Mirella Diaz-Santos
- Department of Psychiatry and Biobehavioral Sciences, University of California, 760 Westwood Plaza, Suite 28-224, Los Angeles, 90095, USA
| | - Spencer Frei
- Department of Psychiatry and Biobehavioral Sciences, University of California, 760 Westwood Plaza, Suite 28-224, Los Angeles, 90095, USA.,Department of Statistics, University of California, Los Angeles, USA
| | - Bianca H Dang
- Department of Psychiatry and Biobehavioral Sciences, University of California, 760 Westwood Plaza, Suite 28-224, Los Angeles, 90095, USA
| | - Pashmeen Kaur
- Department of Statistics, University of California, Los Angeles, USA.,Department of Statistics, Ohio State University, Columbus, USA
| | - Patrick Lyden
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Richard Buxton
- Department of Radiology, University of California, San Diego, USA
| | - Pamela K Douglas
- Department of Psychiatry and Biobehavioral Sciences, University of California, 760 Westwood Plaza, Suite 28-224, Los Angeles, 90095, USA.,Institute for Simulation and Training, University of Central Florida, Orlando, USA
| | - Robert M Bilder
- Department of Psychiatry and Biobehavioral Sciences, University of California, 760 Westwood Plaza, Suite 28-224, Los Angeles, 90095, USA
| | | | - Karl J Friston
- Institute of Neurology, University College London, London, UK
| | - Usha Nookala
- Department of Psychiatry and Biobehavioral Sciences, University of California, 760 Westwood Plaza, Suite 28-224, Los Angeles, 90095, USA
| | - Susan Y Bookheimer
- Department of Psychiatry and Biobehavioral Sciences, University of California, 760 Westwood Plaza, Suite 28-224, Los Angeles, 90095, USA
| |
Collapse
|
14
|
Wroolie T, Roat-Shumway S, Watson K, Reiman E, Rasgon N. Effects of LDL Cholesterol and Statin Use on Verbal Learning and Memory in Older Adults at Genetic Risk for Alzheimer's Disease. J Alzheimers Dis 2020; 75:903-910. [PMID: 32390619 DOI: 10.3233/jad-191090] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND The apolipoprotein epsilon 4 (APOE4) allele is a well-established genetic risk factor for Alzheimer's disease (AD). However, there are mixed findings as to how the APOE4 allele modifies the effects of both higher low-density lipoprotein cholesterol (LDL) and statin use on cognitive functioning. OBJECTIVE This study sought to examine the effects of LDL levels and statin use on verbal learning and memory, as modified by the presence of the APOE4 allele, in a sample of cognitively unimpaired, older adults at risk for AD. METHODS Neuropsychological, LDL, statin use, and APOE4 data were extracted from an ongoing longitudinal study at the Banner Alzheimer's Institute in Arizona. Participants were cognitively unimpaired based on Mini-Mental State Examination scores within a normal range, aged 47-75, with a family history of probable AD in at least one first-degree relative. RESULTS In the whole sample, higher LDL was associated with worse immediate verbal memory in APOE4 non-carriers, but did not have an effect on immediate verbal memory in APOE4 carriers. In APOE4 non-carriers, statin use was associated with better verbal learning, but did not have an effect on verbal learning in APOE4 carriers. Among women, higher LDL in APOE4 carriers was associated with worse verbal learning than in APOE4 non-carriers, and statin use in APOE4 non-carriers was associated with better verbal learning and immediate and delayed verbal memory but worse performances on these tasks in APOE4 carriers. CONCLUSION LDL and statin use may have differential effects on verbal learning and/or memory depending on genetic risk for AD. Women appear to be particularly vulnerable to statin use depending on their APOE4 status.
Collapse
Affiliation(s)
- Tonita Wroolie
- Department of Psychiatry & Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Siena Roat-Shumway
- Department of Psychiatry & Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Katie Watson
- Department of Psychiatry & Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Eric Reiman
- Banner Alzheimer's Institute, Stead Family Memory Center, Phoenix, AZ, USA
| | - Natalie Rasgon
- Department of Psychiatry & Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
15
|
Causal association of circulating cholesterol levels with dementia: a mendelian randomization meta-analysis. Transl Psychiatry 2020; 10:145. [PMID: 32398686 PMCID: PMC7217910 DOI: 10.1038/s41398-020-0822-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 04/21/2020] [Accepted: 04/28/2020] [Indexed: 12/23/2022] Open
Abstract
Prospective studies have shown that abnormally circulating cholesterol is associated with the risk of dementia. However, whether the association is causal or not remains unclear. We attempt to infer the causal association in a MR meta-analysis by using ApoE gene polymorphisms as instrument variables. Studies with dementia risk (27 studies) or circulating lipid levels (7 studies) were included, with totally 3136 dementia patients and 3103 healthy controls. The analyses showed that carriers of ε2 allele significantly were of decreased risk of AD (OR = 0.70; 95% CI: 0.58-0.84; P < 0.01), whereas carriers of ε4 allele were of increased risk of AD (OR = 3.62; 95% CI: 3.03-4.32; P < 0.05), compared to these of ε3 allele. Circulating TC was significantly reduced in carriers of ε2 allele (WMD = - 0.29 mmol/L; 95% CI: -0.54 to -0.03; P < 0.05) and increased in carriers of ε4 allele (WMD = 0.42 mmol/l; 95% CI: 0.001-0.84; P < 0.05). In addition, carriers of ε4 allele had reduction in circulating HDL-C (WMD = - 0.04 mmol/L; 95% CI: - 0.07 to -0.001; P < 0.05). In comparing allele ε2 with ε3, the predicted OR of having AD for 1 mg/dL increment in circulating TC was 0.97 (95% CI: 0.86-0.98; P < 0.05). Comparing allele ε4 with ε3, the predicted OR for a 1 mg/dL increment in TC was 1.08 (95% CI: 1.05-17.58; P < 0.05), and reduction in HDL-C was 2.30 (95% CI: 1.51-43.99; P < 0.05). Our findings demonstrate that high circulating TC and reduced HDL-C levels might be potential risk factors of the development of AD.
Collapse
|
16
|
John J, Sapa NK, Shenoy RR. Virgin Coconut Oil Ameliorates Colchicine Induced Cognitive Dysfunction- A Preclinical Study. PHARMACEUTICAL SCIENCES 2020. [DOI: 10.34172/ps.2019.61] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background
: Virgin coconut oil (VCO) has been identified as a potential cognitive strengthener associated with Alzheimer’s disease (AD). It contains medium chain fatty acids (MCFA) which are absorbed and easily metabolized by the liver to form ketone bodies. Ketone bodies are converted to acetyl Co-A in the brain which then enters the citric acid cycle to provide ATP and also serves as precursors of acetylcholine in neurons. Sunflower oil (SO) contains poly unsaturated fatty acids which has both anti-inflammatory and neuroprotective actions. To compare the neuroprotective effects of VCO and SO on biochemical parameters involved in the cognitive dysfunction induced by colchicine through intracerebroventricular (i.c.v) route.To assess the role of polyphenols and MCFA present in VCO in preventing oxidative stress and its influence on in neuroprotection and memory enhancement. Methods: In the present study, we induced dementia through i.c.v injection of colchicine after giving the diet enriched VCO and SO in rats for 60 days. Rats were sacrificed on the 22nd day after the administration of colchicine. Behavioral parameters were assessed during the study period and biochemical estimations were performed using frontal cortex and hippocampus isolated from rat brain. Results: From the memory and learning tests by Morris water maze, VCO treated group performed better than SO treated rats. VCO reversed the antagonistic effects induced by colchicine by decreasing the acetylcholinesterase and malondialdehyde levels and increasing the levels of catalase and superoxide dismutase. SO only reduced malondialdehyde levels in cortex and hippocampus. Conclusion: The results demonstrated potential beneficiary effects of VCO in the cognitive dysfunction induced by colchicine by enhancing acetylcholine levels in the frontal cortex and hippocampus and also by reducing oxidative stress induced by physiological oxidants.
Collapse
Affiliation(s)
- Jeena John
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India-576104
| | - Naveen Kumar Sapa
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India-576104
| | - Rekha R Shenoy
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India-576104
| |
Collapse
|
17
|
Williams T, Borchelt DR, Chakrabarty P. Therapeutic approaches targeting Apolipoprotein E function in Alzheimer's disease. Mol Neurodegener 2020; 15:8. [PMID: 32005122 PMCID: PMC6995170 DOI: 10.1186/s13024-020-0358-9] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 01/14/2020] [Indexed: 12/12/2022] Open
Abstract
One of the primary genetic risk factors for Alzheimer’s disease (AD) is the presence of the Ɛ4 allele of apolipoprotein E (APOE). APOE is a polymorphic lipoprotein that is a major cholesterol carrier in the brain. It is also involved in various cellular functions such as neuronal signaling, neuroinflammation and glucose metabolism. Humans predominantly possess three different allelic variants of APOE, termed E2, E3, and E4, with the E3 allele being the most common. The presence of the E4 allele is associated with increased risk of AD whereas E2 reduces the risk. To understand the molecular mechanisms that underlie APOE-related genetic risk, considerable effort has been devoted towards developing cellular and animal models. Data from these models indicate that APOE4 exacerbates amyloid β plaque burden in a dose-dependent manner. and may also enhance tau pathogenesis in an isoform-dependent manner. Other studies have suggested APOE4 increases the risk of AD by mechanisms that are distinct from modulation of Aβ or tau pathology. Further, whether plasma APOE, by influencing systemic metabolic pathways, can also possibly alter CNS function indirectly is not complete;y understood. Collectively, the available studies suggest that APOE may impact multiple signaling pathways and thus investigators have sought therapeutics that would disrupt pathological functions of APOE while preserving or enhancing beneficial functions. This review will highlight some of the therapeutic strategies that are currently being pursued to target APOE4 towards preventing or treating AD and we will discuss additional strategies that holds promise for the future.
Collapse
Affiliation(s)
- Tosha Williams
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA.,Department of Neuroscience, University of Florida, Gainesville, FL, 32610, USA
| | - David R Borchelt
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA.,Department of Neuroscience, University of Florida, Gainesville, FL, 32610, USA.,McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Paramita Chakrabarty
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA. .,Department of Neuroscience, University of Florida, Gainesville, FL, 32610, USA. .,McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
18
|
Elevated cellular cholesterol in Familial Alzheimer's presenilin 1 mutation is associated with lipid raft localization of β-amyloid precursor protein. PLoS One 2019; 14:e0210535. [PMID: 30682043 PMCID: PMC6347419 DOI: 10.1371/journal.pone.0210535] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 12/26/2018] [Indexed: 11/30/2022] Open
Abstract
Familial Alzheimer’s disease (FAD)-associated presenilin 1 (PS1) serves as a catalytic subunit of γ-secretase complex, which mediates the proteolytic liberation of β-amyloid (Aβ) from β-amyloid precursor protein (APP). In addition to its proteolytic role, PS1 is involved in non-proteolytic functions such as protein trafficking and ion channel regulation. Furthermore, postmortem AD brains as well as AD patients showed dysregulation of cholesterol metabolism. Since cholesterol has been implicated in regulating Aβ production, we investigated whether the FAD PS1-associated cholesterol elevation could influence APP processing. We found that in CHO cells stably expressing FAD-associated PS1 ΔE9, total cholesterol levels are elevated compared to cells expressing wild-type PS1. We also found that localization of APP in cholesterol-enriched lipid rafts is substantially increased in the mutant cells. Reducing the cholesterol levels by either methyl-β-cyclodextrin or an inhibitor of CYP51, an enzyme mediating the elevated cholesterol in PS1 ΔE9-expressing cells, significantly reduced lipid raft-associated APP. In contrast, exogenous cholesterol increased lipid raft-associated APP. These data suggest that in the FAD PS1 ΔE9 cells, the elevated cellular cholesterol level contributes to the altered APP processing by increasing APP localized in lipid rafts.
Collapse
|
19
|
Ostroumova OD, Chikh EV, Rebrova EV, Ryazanova AY. Statin Therapy and Cognitive Impairment: Benefit or Harm? RATIONAL PHARMACOTHERAPY IN CARDIOLOGY 2018. [DOI: 10.20996/1819-6446-2018-14-4-529-536] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Statins are now widely used drugs for the treatment of dyslipidemia, effective drugs for lowering the level of low-density lipoprotein cholesterol, and also for reducing the risk of cardiovascular and cerebrovascular events. It is believed that statins are well tolerated. However, the potential relationship between statins and cognitive impairment in some people is assumed. This review paper was written in the light of the search for information on a specific problem of the potential adverse effects of statins on the cognitive function. The purpose of the article is to seek advice for health professionals on monitoring and reducing the risk of potential cognitive impairment during statin therapy. Rosuvastatin may be safer amongst statins in terms of influencing cognitive function. Evaluation of cognitive dysfunction in patients before starting therapy with statins is not necessary. Therapy with statins is not accompanied by a risk of developing cognitive dysfunction according to cohort and randomized studies. The presence of cognitive dysfunction and the exclusion of other possible causes of it, as well as the evaluation of the benefit/risk ratio for the abolition of statin therapy, are necessary in detecting cognitive dysfunction during statin therapy. A decrease in the dose of statin or the cessation of its use to assess the reversibility of symptoms is possible on the basis of the individual characteristics of the patient. Alternative replacement for another inhibitor of HMG-CoA reductase should occur if the statin is discontinued. A drug that less penetrates the blood-brain barrier, for example rosuvastatin, is more preferable.
Collapse
|
20
|
Swanson A, Wolf T, Sitzmann A, Willette AA. Neuroinflammation in Alzheimer's disease: Pleiotropic roles for cytokines and neuronal pentraxins. Behav Brain Res 2018; 347:49-56. [PMID: 29462653 PMCID: PMC5988985 DOI: 10.1016/j.bbr.2018.02.015] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 01/30/2018] [Accepted: 02/13/2018] [Indexed: 12/14/2022]
Abstract
Neuroinflammation is a potential factor speculated to underlie Alzheimer's disease (AD) etiopathogenesis and progression. The overwhelming focus in this area of research to date has been on the chronic upregulation of pro-inflammatory cytokines to understand how neuroinflammatory mechanisms contribute to neurodegeneration. Yet, it is important to understand the pleiotropic roles of these cytokines in modulating neuroinflammation in which they cannot be labeled as a strictly "good" or "bad" biomarker phenotype. As such, biomarkers with more precise functions are needed to better understand how neuroinflammation impacts the brain in AD. Neuronal pentraxins are a concentration- dependent group of pro- or anti- inflammatory cytokines. There is contradictory evidence of these pentraxins as being both neuroprotective and potentially detrimental in AD. Potential neuroprotective examples include their ability to predict AD-related outcomes such as cognition, memory function and synaptic refinement. This review will briefly outline the basis of AD and subsequently summarize findings for neuropathological mechanisms of neuroinflammation, roles for traditional pro-and anti-inflammatory cytokines, and data found thus far on the neuronal pentraxins.
Collapse
Affiliation(s)
- Ashley Swanson
- Department of Food Science and Human Nutrition, Iowa State University, 2312 Food Sciences Building, 536 Farm House Lane, Ames, IA 50011, United States.
| | - Tovah Wolf
- Department of Food Science and Human Nutrition, Iowa State University, 2312 Food Sciences Building, 536 Farm House Lane, Ames, IA 50011, United States.
| | - Alli Sitzmann
- Department of Psychology, Iowa State University, W112 Lagomarcino Hall, 901 Stange Road, Ames, IA 50011, United States.
| | - Auriel A Willette
- Department of Food Science and Human Nutrition, Iowa State University, 2312 Food Sciences Building, 536 Farm House Lane, Ames, IA 50011, United States; Department of Psychology, Iowa State University, W112 Lagomarcino Hall, 901 Stange Road, Ames, IA 50011, United States; Department of Biomedical Sciences, Iowa State University, 2008 Veterinary Medicine, Ames, IA 50011, United States; Department of Neurology, University of Iowa, 2007 Roy Carver Pavilion, 200 Hawkins Drive, Iowa City, IA 52242, United States.
| |
Collapse
|
21
|
Huang X, Dong S, Zhen J, Zhang H, Lin T, Zeng Y, Van Halm-Lutterodt N, Yuan L. The Role of ApoE Polymorphism in the Relationship between Serum Steroid Hormone Levels and Cognition in Older Chinese Adults: A Cross-Sectional Study. Front Endocrinol (Lausanne) 2018; 9:71. [PMID: 29559956 PMCID: PMC5845719 DOI: 10.3389/fendo.2018.00071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 02/16/2018] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Epidemiology studies have indicated an association of apolipoprotein E (ApoE) genetic polymorphism and circulating steroid hormone levels with the risk of Alzheimer's disease. The established physiologic relationship between apolipoproteins and steroid hormone indicate an important role of ApoE polymorphism in impacting the relationship between serum steroid hormones and cognition in the elderly. STUDY DESIGN A total of 500 Chinese adults aged between 50 and 75 participated in this community-based cross-sectional study. Blood samples were collected in the morning for ApoE genotyping and serum parameter assessment. Cognitive performance of participants was evaluated by Montreal Cognitive Assessment test. RESULTS Age, gender, educational level, smoking, and physical activity levels are factors associated with cognitive performance in this older Chinese adults. Compared to the control subjects, MCI subjects demonstrated higher serum total cholesterol, HDL-C, and estradiol status (P < 0.05). ApoE genotype difference of serum lipid profile was observed with a relatively higher mean serum triglyceride levels in ApoE2 and ApoE4 carriers (P < 0.05), and lower mean serum HDL-C level in ApoE4 carriers (P < 0.05). Memory and delayed recall ability was serum estradiol level related; and subjects with higher circulating estradiol concentration exhibited lower memory and delayed recall ability (P < 0.05). The association of serum estradiol and cortisol concentration with cognitive performance was ApoE genotypes dependent. Poor cognitive performance was observed in ApoE2 and ApoE4 carriers with higher serum estradiol level (P < 0.05). Moreover, ApoE2 and ApoE4 carriers with higher serum cortisol status demonstrated decreased language ability (P < 0.05). Multiple logistic regression analysis indicates that subjects with higher serum estradiol status may have an increased risk for MCI [OR = 2.004, 95% confidence interval (CI): 1.135, 3.540; P = 0.017]. ApoE2 carriers with higher serum steroid levels may be potentially predisposed to an increased risk of MCI (OR = 3.353; 95% CI: 1.135, 9.907; P = 0.029). CONCLUSION Cognitive outcomes in older Chinese adults are associated with serum steroid hormone status. Higher serum steroid levels in ApoE2 carriers might pose an increased risk of MCI in the elderly.
Collapse
Affiliation(s)
- Xiaochen Huang
- School of Public Health, Capital Medical University, Beijing, China
| | - Shengqi Dong
- School of Public Health, Capital Medical University, Beijing, China
| | - Jie Zhen
- School of Public Health, Capital Medical University, Beijing, China
| | - Huiqiang Zhang
- School of Public Health, Capital Medical University, Beijing, China
| | - Tong Lin
- School of Public Health, Capital Medical University, Beijing, China
| | - Yuhong Zeng
- School of Public Health, Capital Medical University, Beijing, China
| | - Nicholas Van Halm-Lutterodt
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Keck Medical Center of USC, Department of Orthopaedics and Neurosurgery, University of Southern California, Los Angeles, CA, United States
| | - Linhong Yuan
- School of Public Health, Capital Medical University, Beijing, China
- *Correspondence: Linhong Yuan,
| |
Collapse
|
22
|
Medina M, Khachaturian ZS, Rossor M, Avila J, Cedazo-Minguez A. Toward common mechanisms for risk factors in Alzheimer's syndrome. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2017; 3:571-578. [PMID: 29124116 PMCID: PMC5671628 DOI: 10.1016/j.trci.2017.08.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The global strategic goal of reducing health care cost, especially the prospects for massive increases due to expanding markets for health care services demanded by aging populations and/or people with a wide range of chronic disorders-disabilities, is a complex and formidable challenge with many facets. Current projections predict marked increases in the demand for health driven by both the exponential climb in the prevalence of chronic disabilities and the increases in the absolute numbers of people in need of some form of health care. Thus, the looming predicament for the economics of health care systems worldwide mandates the formulation of a strategic goal to foster significant expansion of global R&D efforts to discover and develop wide-ranging interventions to delay and/or prevent the onset of chronic disabling conditions. The rationale for adopting such a tactical objective is based on the premise that the costs and prevalence of chronic disabling conditions will be reduced by half even if a modest delay of 5 years in the onset of disability is obtained by a highly focused multinational research initiative. Because of the recent history of many failures in drug trials, the central thesis of this paper is to argue for the exploration-adoption of novel mechanistic ideas, theories, and paradigms for developing wide range and/or types of interventions. Although the primary focus of our discussion has been on biological approaches to therapy, we recognize the importance of emerging knowledge on nonpharmacological interventions and their potential impact in reducing health care costs. Although we may not find a drug to cure or prevent dementia for a long time, research is starting to demonstrate the potential contributes of nonpharmacological interventions toward the economics of health care in terms of rehabilitation, promoting autonomy, and potential to delay institutionalization, thus promoting healthy aging and reductions in the cost of care.
Collapse
Affiliation(s)
- Miguel Medina
- CIBERNED (Network Center for Biomedical Research in Neurodegenerative Diseases), ISCIII, Madrid, Spain
- CIEN Foundation, Reina Sofia Foundation Alzheimer Center, Madrid, Spain
| | | | - Martin Rossor
- Institute of Neurology, University College London, London, UK
| | - Jesús Avila
- CIBERNED (Network Center for Biomedical Research in Neurodegenerative Diseases), ISCIII, Madrid, Spain
- Neurobiology Laboratory, Center for Molecular Biology “Severo Ochoa” CSIC-UAM, Madrid, Spain
| | - Angel Cedazo-Minguez
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
23
|
Hypercholesterolaemia and vascular dementia. Clin Sci (Lond) 2017; 131:1561-1578. [PMID: 28667059 DOI: 10.1042/cs20160382] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Revised: 03/08/2017] [Accepted: 03/21/2017] [Indexed: 12/30/2022]
Abstract
Vascular dementia (VaD) is the second commonest cause of dementia. Stroke is the leading cause of disability in adults in developed countries, the second major cause of dementia and the third commonest cause of death. Traditional vascular risk factors-diabetes, hypercholesterolaemia, hypertension and smoking-are implicated as risk factors for VaD. The associations between cholesterol and small vessel disease (SVD), stroke, cognitive impairment and subsequent dementia are complex and as yet not fully understood. Similarly, the effects of lipids and lipid-lowering therapy on preventing or treating dementia remain unclear; the few trials that have assessed lipid-lowering therapy for preventing (two trials) or treating (four trials) dementia found no evidence to support the use of lipid-lowering therapy for these indications. It is appropriate to treat those patients with vascular risk factors that meet criteria for lipid-lowering therapy for the primary and secondary prevention of cardiovascular and cerebrovascular events, and in line with current guidelines. Managing the individual patient in a holistic manner according to his or her own vascular risk profile is recommended. Although the paucity of randomized controlled evidence makes for challenging clinical decision making, it provides multiple opportunities for on-going and future research, as discussed here.
Collapse
|
24
|
Geifman N, Brinton RD, Kennedy RE, Schneider LS, Butte AJ. Evidence for benefit of statins to modify cognitive decline and risk in Alzheimer's disease. Alzheimers Res Ther 2017; 9:10. [PMID: 28212683 PMCID: PMC5316146 DOI: 10.1186/s13195-017-0237-y] [Citation(s) in RCA: 140] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 01/23/2017] [Indexed: 01/07/2023]
Abstract
BACKGROUND Despite substantial research and development investment in Alzheimer's disease (AD), effective therapeutics remain elusive. Significant emerging evidence has linked cholesterol, β-amyloid and AD, and several studies have shown a reduced risk for AD and dementia in populations treated with statins. However, while some clinical trials evaluating statins in general AD populations have been conducted, these resulted in no significant therapeutic benefit. By focusing on subgroups of the AD population, it may be possible to detect endotypes responsive to statin therapy. METHODS Here we investigate the possible protective and therapeutic effect of statins in AD through the analysis of datasets of integrated clinical trials, and prospective observational studies. RESULTS Re-analysis of AD patient-level data from failed clinical trials suggested by trend that use of simvastatin may slow the progression of cognitive decline, and to a greater extent in ApoE4 homozygotes. Evaluation of continual long-term use of various statins, in participants from multiple studies at baseline, revealed better cognitive performance in statin users. These findings were supported in an additional, observational cohort where the incidence of AD was significantly lower in statin users, and ApoE4/ApoE4-genotyped AD patients treated with statins showed better cognitive function over the course of 10-year follow-up. CONCLUSIONS These results indicate that the use of statins may benefit all AD patients with potentially greater therapeutic efficacy in those homozygous for ApoE4.
Collapse
Affiliation(s)
- Nophar Geifman
- The Manchester Molecular Pathology Innovation Centre, University of Manchester, 3rd Floor Citylabs, Nelson St, Manchester, M13 9NQ UK
- Health eResearch Centre, Division of Informatics, Imaging & Data Sciences, University of Manchester, Manchester, UK
| | - Roberta Diaz Brinton
- Center for Innovation in Brain Science, School of Medicine, Departments of Pharmacology and Neurology, University of Arizona, Tucson, AZ USA
| | - Richard E. Kennedy
- School of Medicine, University of Alabama at Birmingham, Birmingham, AL USA
| | - Lon S. Schneider
- Keck School of Medicine, University of Southern California, Los Angeles, CA USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA USA
| | - Atul J. Butte
- Institute for Computational Health Sciences, University of California San Francisco, San Francisco, CA USA
| |
Collapse
|
25
|
Loke SY, Wong PTH, Ong WY. Global gene expression changes in the prefrontal cortex of rabbits with hypercholesterolemia and/or hypertension. Neurochem Int 2016; 102:33-56. [PMID: 27890723 DOI: 10.1016/j.neuint.2016.11.010] [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: 02/17/2016] [Revised: 11/14/2016] [Accepted: 11/23/2016] [Indexed: 02/01/2023]
Abstract
Although many studies have identified a link between hypercholesterolemia or hypertension and cognitive deficits, till date, comprehensive gene expression analyses of the brain under these conditions is still lacking. The present study was carried out to elucidate differential gene expression changes in the prefrontal cortex (PFC) of New Zealand white rabbits exposed to hypercholesterolemia and/or hypertension with a view of identifying gene networks at risk. Microarray analyses of the PFC of hypercholesterolemic rabbits showed 850 differentially expressed genes (DEGs) in the cortex of hypercholesterolemic rabbits compared to controls, but only 5 DEGs in hypertensive rabbits compared to controls. Up-regulated genes in the PFC of hypercholesterolemic rabbits included CIDEC, ODF2, RNASEL, FSHR, CES3 and MAB21L3, and down-regulated genes included FAM184B, CUL3, LOC100351029, TMEM109, LOC100357097 and PFDN5. Comparison with our previous study on the middle cerebral artery (MCA) of the same rabbits showed many differentially expressed genes in common between the PFC and MCA, during hypercholesterolemia. Moreover, these genes tended to fall into the same functional networks, as revealed by IPA analyses, with many identical node molecules. These include: proteasome, insulin, Akt, ERK1/2, histone, IL12, interferon alpha and NFκB. Of these, PSMB4, PSMD4, PSMG1 were chosen as representatives of genes related to the proteasome for verification by quantitative RT-PCR. Results indicate significant downregulation of all three proteasome associated genes in the PFC. Immunostaining showed significantly increased number of Aβ labelled cells in layers III and V of the cortex after hypercholesterolemia and hypertension, which may be due to decreased proteasome activity and/or increased β- or γ-secretase activity. Knowledge of altered gene networks during hypercholesterolemia and/or hypertension could inform our understanding of the link between these conditions and cognitive deficits in vascular dementia or Alzheimer's disease.
Collapse
Affiliation(s)
- Sau-Yeen Loke
- Department of Anatomy, National University of Singapore, 119260, Singapore
| | - Peter Tsun-Hon Wong
- Department of Pharmacology, National University of Singapore, 119260, Singapore
| | - Wei-Yi Ong
- Department of Anatomy, National University of Singapore, 119260, Singapore; Neurobiology and Ageing Research Program, Life Sciences Institute, National University of Singapore, 119260, Singapore.
| |
Collapse
|
26
|
Hendrie HC. Exploration of Environmental and Genetic Risk Factors for Alzheimer's Disease: The Value of Cross-Cultural Studies. CURRENT DIRECTIONS IN PSYCHOLOGICAL SCIENCE 2016. [DOI: 10.1111/1467-8721.00125] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Advances in molecular genetics have revolutionized epidemiological research. It is now possible to combine the techniques of population genetics with research on risk factors to construct genetic-environmental interactive models that explain geographic-ethnic variations in disease rates. Cross-cultural studies involving populations from developing and developed countries offer a unique opportunity for constructing these models by providing a wide diversity of environmental exposures. Results from a comparative Indianapolis-Ibadan study suggest that Alzheimer's disease incidence rates are lower in Yoruba than in African Americans and that these lower rates may be due to a combination of genetic and environmental, primarily dietary, influences.
Collapse
Affiliation(s)
- Hugh C. Hendrie
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
27
|
Riedel BC, Thompson PM, Brinton RD. Age, APOE and sex: Triad of risk of Alzheimer's disease. J Steroid Biochem Mol Biol 2016; 160:134-47. [PMID: 26969397 PMCID: PMC4905558 DOI: 10.1016/j.jsbmb.2016.03.012] [Citation(s) in RCA: 420] [Impact Index Per Article: 46.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2015] [Revised: 03/02/2016] [Accepted: 03/06/2016] [Indexed: 02/06/2023]
Abstract
Age, apolipoprotein E ε4 (APOE) and chromosomal sex are well-established risk factors for late-onset Alzheimer's disease (LOAD; AD). Over 60% of persons with AD harbor at least one APOE-ε4 allele. The sex-based prevalence of AD is well documented with over 60% of persons with AD being female. Evidence indicates that the APOE-ε4 risk for AD is greater in women than men, which is particularly evident in heterozygous women carrying one APOE-ε4 allele. Paradoxically, men homozygous for APOE-ε4 are reported to be at greater risk for mild cognitive impairment and AD. Herein, we discuss the complex interplay between the three greatest risk factors for Alzheimer's disease, age, APOE-ε4 genotype and chromosomal sex. We propose that the convergence of these three risk factors, and specifically the bioenergetic aging perimenopause to menopause transition unique to the female, creates a risk profile for AD unique to the female. Further, we discuss the specific risk of the APOE-ε4 positive male which appears to emerge early in the aging process. Evidence for impact of the triad of AD risk factors is most evident in the temporal trajectory of AD progression and burden of pathology in relation to APOE genotype, age and sex. Collectively, the data indicate complex interactions between age, APOE genotype and gender that belies a one size fits all approach and argues for a precision medicine approach that integrates across the three main risk factors for Alzheimer's disease.
Collapse
Affiliation(s)
- Brandalyn C Riedel
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA 90089, USA
| | - Paul M Thompson
- USC Institute for Neuroimaging and Informatics, University of Southern California, Marina del Rey, CA 90292, USA
| | - Roberta Diaz Brinton
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
28
|
McGuinness B, Craig D, Bullock R, Passmore P, Cochrane Dementia and Cognitive Improvement Group. Statins for the prevention of dementia. Cochrane Database Syst Rev 2016; 2016:CD003160. [PMID: 26727124 PMCID: PMC9346344 DOI: 10.1002/14651858.cd003160.pub3] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BACKGROUND This is an update of a Cochrane review first published in 2001 and then updated in 2009. Vascular risk factors including high cholesterol levels increase the risk of dementia due to Alzheimer's disease and of vascular dementia. Some observational studies have suggested an association between statin use and lowered incidence of dementia. OBJECTIVES To evaluate the efficacy and safety of statins for the prevention of dementia in people at risk of dementia due to their age and to determine whether the efficacy and safety of statins for this purpose depends on cholesterol level, apolipoprotein E (ApoE) genotype or cognitive level. SEARCH METHODS We searched ALOIS (the Specialized Register of the Cochrane Dementia and Cognitive Improvement Group), The Cochrane Library, MEDLINE, EMBASE, PsycINFO, CINAHL, LILACS, ClinicalTrials.gov and the World Health Organization (WHO) Portal on 11 November 2015. SELECTION CRITERIA We included double-blind, randomised, placebo-controlled trials in which statins were administered for at least 12 months to people at risk of dementia. DATA COLLECTION AND ANALYSIS We used standard methodological procedures expected by Cochrane. MAIN RESULTS We included two trials with 26,340 participants aged 40 to 82 years of whom 11,610 were aged 70 or older. All participants had a history of, or risk factors for, vascular disease. The studies used different statins (simvastatin and pravastatin). Mean follow-up was 3.2 years in one study and five years in one study. The risk of bias was low. Only one study reported on the incidence of dementia (20,536 participants, 31 cases in each group; odds ratio (OR) 1.00, 95% confidence interval (CI) 0.61 to 1.65, moderate quality evidence, downgraded due to imprecision). Both studies assessed cognitive function, but at different times using different scales, so we judged the results unsuitable for a meta-analysis. There were no differences between statin and placebo groups on five different cognitive tests (high quality evidence). Rates of treatment discontinuation due to non-fatal adverse events were less than 5% in both studies and there was no difference between statin and placebo groups in the risk of withdrawal due to adverse events (26,340 participants, 2 studies, OR 0.94, 95% CI 0.83 to 1.05). AUTHORS' CONCLUSIONS There is good evidence that statins given in late life to people at risk of vascular disease do not prevent cognitive decline or dementia. Biologically, it seems feasible that statins could prevent dementia due to their role in cholesterol reduction and initial evidence from observational studies was very promising. However, indication bias may have been a factor in these studies and the evidence from subsequent RCTs has been negative. There were limitations in the included studies involving the cognitive assessments used and the inclusion of participants at moderate to high vascular risk only.
Collapse
Affiliation(s)
- Bernadette McGuinness
- Queen's University BelfastCentre for Public HealthInstitute of Clinical Sciences, Block BGrosvenor RoadBelfastCo AntrimUKBT12 6BA
| | - David Craig
- Craigavon Area HospitalGeriatric MedicineCraigavonNorthern IrelandUK
| | - Roger Bullock
- Kingshill Research Centre, Victoria HospitalOkus RoadSwindonUKSN4 4HZ
| | - Peter Passmore
- Queen's University BelfastCentre for Public HealthInstitute of Clinical Sciences, Block BGrosvenor RoadBelfastCo AntrimUKBT12 6BA
| | | |
Collapse
|
29
|
Agarwal R, Talwar P, Kushwaha SS, Tripathi CB, Kukreti R. Effect of apolipoprotein E (APO E) polymorphism on leptin in Alzheimer's disease. Ann Indian Acad Neurol 2015; 18:320-6. [PMID: 26425011 PMCID: PMC4564468 DOI: 10.4103/0972-2327.157255] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background: Leptin, a 16 kDa peptide hormone synthesized and secreted specifically from white adipose cells protects neurons against amyloid β-induced toxicity, by increasing Apolipoprotein E (APO E)-dependent uptake of β amyloid into the cells, thereby, protect individuals from developing Alzheimer's disease (AD). The APO E ε4 allele is a known genetic risk factor for AD by accelerating onset. It is estimated that the lifetime risk of developing AD increases to 29% for carriers with one ε4 allele and 9% for those with no ε4 allele. Objectives: To determine the levels of serum leptin, cholesterol, low density lipoprotein (LDL-C), and high density lipoprotein (HDL-C) in the diagnosed cases of AD and the association of them with cognitive decline and Apolipoprotein E (APO E) genotypes in AD. Materials and Methods: Serum levels of serum leptin, cholesterol, LDL-C, and HDL-C along with APO E polymorphism were studied in 39 subjects with probable AD and 42 cognitive normal individuals. Results: AD group showed significantly lower levels of leptin (P = 0.00) as compared to control group. However, there was no significant difference in cholesterol, triglycerides, LDL-C, and HDL-C levels in AD and control groups. The frequency of ε4 allele in AD (38.5%) was found to be significantly higher than in control (10.3%). ε3 allele was more frequent than ε4 allele in AD and control group.
Collapse
Affiliation(s)
- Rachna Agarwal
- Department of Neurochemistry, Institute of Human Behaviour and Allied Sciences, Delhi, India
| | - Puneet Talwar
- Genetics and Molecular Medicine, Institute of Genomics and Integrated Biology, Delhi, India
| | - Suman S Kushwaha
- Department of Neurology, Institute of Human Behaviour and Allied Sciences, Delhi, India
| | | | - Ritushree Kukreti
- Genetics and Molecular Medicine, Institute of Genomics and Integrated Biology, Delhi, India
| |
Collapse
|
30
|
Molecular dynamics simulation study reveals potential substrate entry path into γ-secretase/presenilin-1. J Struct Biol 2015; 191:120-9. [DOI: 10.1016/j.jsb.2015.07.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 05/29/2015] [Accepted: 07/01/2015] [Indexed: 11/20/2022]
|
31
|
John J, Nampoothiri M, Kumar N, Mudgal J, Nampurath GK, Chamallamudi MR. Sesamol, a lipid lowering agent, ameliorates aluminium chloride induced behavioral and biochemical alterations in rats. Pharmacogn Mag 2015; 11:327-36. [PMID: 25829772 PMCID: PMC4378131 DOI: 10.4103/0973-1296.153086] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2014] [Revised: 08/10/2014] [Accepted: 03/12/2015] [Indexed: 01/01/2023] Open
Abstract
Background: Sesame oil from the seeds of Sesamum indicum Linn. (Pedaliaceae) has been used traditionally in Indian medical practice of Ayurveda in the treatment of central nervous system disorders and insomnia. A few published reports favor the anti-dementia effect of sesamol (SML), an active constituent of sesame oil. Objective: Thus, the present study was aimed to explore the anti-dementia effect and possible mechanism (s) of SML in aluminium chloride (AlCl3)-induced cognitive dysfunction model in rodents with special emphasis on memory centers viz., hippocampus and frontal cortex. Methods: Male Wistar rats were exposed to AlCl3 (175 mg/kg p.o.) for 60 days. SML (10 and 20 mg/kg) and rivastigmine (1 mg/kg) were administered orally 45 min before administration of AlCl3 for 60 days. Spatial memory was assessed using Morris water maze test. After 60 days of treatment animals were sacrificed, hippocampus and frontal cortex were collected and analyzed for acetylcholinesterase (AChE) activity, tumor necrosis factor (TNF-α) level, antioxidant enzymes (Glutathione, catalase), lipid peroxidation, and nitrite level. The circulating triglycerides, total cholesterol, low-density lipoprotein (LDL) and high-density lipoprotein (HDL) levels were also analyzed. Results: SML significantly prevented behavioral impairments in aluminium-exposed rats. Treatment with SML reversed the increased cholesterol, triglycerides and LDL while raised the HDL levels. SML significantly corrected the effect of AlCl3 on AChE activity. Further, SML reversed the elevated nitric oxide, TNF-α and reduced antioxidant enzymes in hippocampus and frontal cortex. Conclusion: The present study suggests the neuro-protection by SML against cognitive dysfunction induced by environmental toxin (AlCl3) in hippocampus and frontal cortex.
Collapse
Affiliation(s)
- Jessy John
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal University, Manipal, Karnataka, India
| | - Madhavan Nampoothiri
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal University, Manipal, Karnataka, India
| | - Nitesh Kumar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal University, Manipal, Karnataka, India
| | - Jayesh Mudgal
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal University, Manipal, Karnataka, India
| | - Gopalan Kutty Nampurath
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal University, Manipal, Karnataka, India
| | - Mallikarjuna Rao Chamallamudi
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal University, Manipal, Karnataka, India
| |
Collapse
|
32
|
Wu CK, Yang YH, Lin TT, Tsai CT, Hwang JJ, Lin JL, Chen PC, Chiang FT, Lin LY. Statin use reduces the risk of dementia in elderly patients: a nationwide data survey and propensity analysis. J Intern Med 2015; 277:343-352. [PMID: 24766342 DOI: 10.1111/joim.12262] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVES The objective of this study was to examine the association between the use of statins and the risk of newly diagnosed dementia in an elderly population. DESIGN, SETTING AND PARTICIPANTS Random samples of 1,000,000 individuals covered by the National Health Insurance in Taiwan were included in the analysis. All participants were 65 years or older without dementia and either did or did not start treatment with statins from 1 August 1997 to 31 December 2010. Patients with established dementia before the start of treatment were excluded. Baseline characteristics were matched (by propensity score) in those who did and did not receive statins. RESULTS A total of 57,669 subjects were included in the analysis with approximately 12 years of follow-up. Propensity score matching identified 2003 patients who received statins and another 2003 patients who did not with comparable baseline characteristics. Adjusted hazard ratios (HRs) for dementia were significantly inversely associated with total or daily equivalent statin dosage (total accumulated dose: HRs 0.829, 0.720 and 0.385 from T1 to T3 vs. control, P < 0.001 for trend; mean daily dose: HRs 0.667, 0.798 and 0.503 from T1 to T3 vs. control, P < 0.001). The results remained robust after propensity adjustment. CONCLUSION Independent of traditional risk factors, there was a decrease in newly diagnosed cases of dementia in elderly patients who had received a high total or daily dose of statins. The more potent statins (e.g. atorvastatin and rosuvastatin) seemed to be particularly effective in the prevention of dementia.
Collapse
Affiliation(s)
- C-K Wu
- Division of Cardiology, Department of Internal Medicine, National Taiwan University College of Medicine and Hospital, Taipei, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Y-H Yang
- Department for Traditional Chinese Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan.,Institute of Occupational Medicine and Industrial Hygiene, National Taiwan University College of Public Health, Taipei, Taiwan
| | - T-T Lin
- Division of Cardiology, Department of Internal Medicine, National Taiwan University College of Medicine and Hospital, Taipei, Taiwan
| | - C-T Tsai
- Division of Cardiology, Department of Internal Medicine, National Taiwan University College of Medicine and Hospital, Taipei, Taiwan
| | - J-J Hwang
- Division of Cardiology, Department of Internal Medicine, National Taiwan University College of Medicine and Hospital, Taipei, Taiwan
| | - J-L Lin
- Division of Cardiology, Department of Internal Medicine, National Taiwan University College of Medicine and Hospital, Taipei, Taiwan
| | - P-C Chen
- Institute of Occupational Medicine and Industrial Hygiene, National Taiwan University College of Public Health, Taipei, Taiwan
| | - F-T Chiang
- Division of Cardiology, Department of Internal Medicine, National Taiwan University College of Medicine and Hospital, Taipei, Taiwan.,Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - L-Y Lin
- Division of Cardiology, Department of Internal Medicine, National Taiwan University College of Medicine and Hospital, Taipei, Taiwan
| |
Collapse
|
33
|
Uyrum E, Balbay O, Annakkaya AN, Gulec Balbay E, Silan F, Arbak P. The Relationship between Obstructive Sleep Apnea Syndrome and Apolipoprotein E Genetic Variants. Respiration 2015; 89:195-200. [DOI: 10.1159/000369560] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2013] [Accepted: 11/03/2014] [Indexed: 11/19/2022] Open
|
34
|
A Comparison of Apolipoprotein E Polymorphism in Alzheimer's Disease and Subcortical Vascular Dementia in Koreans. Dement Neurocogn Disord 2015. [DOI: 10.12779/dnd.2015.14.2.70] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
35
|
Abstract
Alzheimer disease (AD) is a dementing, neurodegenerative disorder that affects approximately 500,000 Canadians and its prevalence is expected to double over the next 30 years. Although several medications may temporarily augment cognitive abilities in AD, there presently exists no proven method to avoid the inevitable clinical deterioration in this devastating condition. The delineation of risk factors for the development of AD offers hope for the advent of effective prevention or interventions that might retard the onset of symptoms. In this article, we provide a comprehensive review of midlife risk factors implicated in the etiopathogenesis of sporadic AD. Although some risk factors are heritable and largely beyond our control, others are determined by lifestyle or environment and are potentially modifiable. In a companion paper, we introduce the concept of an Alzheimer Risk Assessment Clinic for ascertainment and mitigation of these and other putative dementia risk factors in middle-aged adults.
Collapse
|
36
|
Wang SH, Huang Y, Yuan Y, Xia WQ, Wang P, Huang R. LDL receptor knock-out mice show impaired spatial cognition with hippocampal vulnerability to apoptosis and deficits in synapses. Lipids Health Dis 2014; 13:175. [PMID: 25413784 PMCID: PMC4258039 DOI: 10.1186/1476-511x-13-175] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 11/11/2014] [Indexed: 11/30/2022] Open
Abstract
Background Evidence from clinical studies support the fact that abnormal cholesterol metabolism in the brain leads to progressive cognitive dysfunction. The low-density lipoprotein receptor (LDLR) is well-known for its role in regulating cholesterol metabolism. Whether LDLR involved in this impaired cognition and the potential mechanisms that underlie this impairment are unknown. Methods Twelve-month-old Ldlr-/- mice (n = 10) and wild-type littermates C57BL/6 J (n = 14) were subjected to the Morris water maze test. At 1 week after completion of the behavioural testing, all of the animals were sacrificed for analysis of synaptic and apoptotic markers. Results The plasma cholesterol concentration of Ldlr-/- mice was increased moderately when compared with C57BL/6 J mice (P < 0.05). Behavioural testing revealed that Ldlr-/- mice displayed impaired spatial memory, and moreover, the expression levels of synaptophysin and the number of synaptophysin-immunoreactive presynaptic boutons in the hippocampal CA1 and dentate gyrus were decreased (all P < 0.05). Ultrastructural changes in the dentate gyrus were observed using transmission electron microscopy. Furthermore, apoptosis in the hippocampus of Ldlr-/- mice was revealed based on elevation, at both the mRNA and protein levels, of the ratio of Bax/Bcl-2 expression (all P < 0.05)and an increase in activated-caspase3 protein level (P < 0.05). Conclusion LDLR deficiency contributes to impaired spatial cognition. This most likely occurs via negative effects that promote apoptosis and synaptic deficits in the hippocampus.
Collapse
Affiliation(s)
- Shao-hua Wang
- Department of Endocrinology, ZhongDa Hospital of Southeast University, No,87 DingJiaQiao Road, Nanjing 210009, PR China.
| | | | | | | | | | | |
Collapse
|
37
|
Mandas A, Congiu MG, Abete C, Dessi S, Manconi PE, Musio M, Columbu S, Racugno W. Cognitive decline and depressive symptoms in late-life are associated with statin use: evidence from a population-based study of Sardinian old people living in their own home. Neurol Res 2014; 36:247-54. [PMID: 24512018 DOI: 10.1179/1743132813y.0000000287] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVES This study was designed to provide further insights into the effects of dyslipidemia (Dys-y) and use of statins (St-y) on cognitive functions and mood in older people. METHODS Three hundred and twenty-nine subjects aged > or = 65 years were screened for cognitive dysfunction using mini mental state examination (MMSE). The geriatric depression scale (GDS) was used to detect depression. Interview questionnaires surveyed activities of daily living (ADL) and instrumental ADL (IADL), as well as other functional disabilities. The presence of neutral lipids (NLs) in cytoplasm of peripheral blood mononuclear cells (PBMCs) was determined with the Oil red O (ORO) staining. RESULTS There was no significant difference in MMSE and GDS scores between normal (Dys-n) and Dys-y. However, when Dys-y subjects were divided into St-y and non-statin users (St-n), significant differences emerged in the scores of MMSE and GDS: St-y had lower MMSE and higher GDS than St-n. Multiple correspondence analysis and logistic regression provided further evidence that elderly St-y were much more likely to suffer of cognitive impairment and depression than St-n. Another interesting finding was that the intensity of NL-PBMCs measured by ORO staining was greater in subjects with altered MMSE compared with cognitively normal subjects. In addition St-y had higher ORO score than St-n. DISCUSSION This is an observational study and cannot, therefore, prove a causal relationship between St-y in the elderly and a higher cognitive decline, nevertheless it provides substantial indications that caution should be exercised in the provision of statins in elderly subjects to avoid accelerated memory loss.
Collapse
|
38
|
Papanikolaou N, Pavlopoulos GA, Pafilis E, Theodosiou T, Schneider R, Satagopam VP, Ouzounis CA, Eliopoulos AG, Promponas VJ, Iliopoulos I. BioTextQuest(+): a knowledge integration platform for literature mining and concept discovery. ACTA ACUST UNITED AC 2014; 30:3249-56. [PMID: 25100685 DOI: 10.1093/bioinformatics/btu524] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
SUMMARY The iterative process of finding relevant information in biomedical literature and performing bioinformatics analyses might result in an endless loop for an inexperienced user, considering the exponential growth of scientific corpora and the plethora of tools designed to mine PubMed(®) and related biological databases. Herein, we describe BioTextQuest(+), a web-based interactive knowledge exploration platform with significant advances to its predecessor (BioTextQuest), aiming to bridge processes such as bioentity recognition, functional annotation, document clustering and data integration towards literature mining and concept discovery. BioTextQuest(+) enables PubMed and OMIM querying, retrieval of abstracts related to a targeted request and optimal detection of genes, proteins, molecular functions, pathways and biological processes within the retrieved documents. The front-end interface facilitates the browsing of document clustering per subject, the analysis of term co-occurrence, the generation of tag clouds containing highly represented terms per cluster and at-a-glance popup windows with information about relevant genes and proteins. Moreover, to support experimental research, BioTextQuest(+) addresses integration of its primary functionality with biological repositories and software tools able to deliver further bioinformatics services. The Google-like interface extends beyond simple use by offering a range of advanced parameterization for expert users. We demonstrate the functionality of BioTextQuest(+) through several exemplary research scenarios including author disambiguation, functional term enrichment, knowledge acquisition and concept discovery linking major human diseases, such as obesity and ageing. AVAILABILITY The service is accessible at http://bioinformatics.med.uoc.gr/biotextquest. CONTACT g.pavlopoulos@gmail.com or georgios.pavlopoulos@esat.kuleuven.be SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Nikolas Papanikolaou
- Division of Basic Sciences, University of Crete, Medical School, Heraklion 71110, Greece, Institute of Marine Biology, Biotechnology and Aquaculture (IMBBC), Hellenic Centre for Marine Research (HCMR), Heraklion, Greece, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Campus Belval, 7, avenue des Hauts-Fourneaux, L-4362 Esch sur Alzette, Luxembourg, Biological Computation & Process Laboratory (BCPL), Chemical Process & Energy Resources Institute (CPERI), Centre for Research & Technology Hellas (CERTH), PO Box 361, GR-57001 Thessalonica, Greece, Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, 70013 Heraklion, Crete, Greece and Department of Biological Sciences, Bioinformatics Research Laboratory, University of Cyprus, PO Box 20537, CY 1678, Nicosia, Cyprus
| | - Georgios A Pavlopoulos
- Division of Basic Sciences, University of Crete, Medical School, Heraklion 71110, Greece, Institute of Marine Biology, Biotechnology and Aquaculture (IMBBC), Hellenic Centre for Marine Research (HCMR), Heraklion, Greece, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Campus Belval, 7, avenue des Hauts-Fourneaux, L-4362 Esch sur Alzette, Luxembourg, Biological Computation & Process Laboratory (BCPL), Chemical Process & Energy Resources Institute (CPERI), Centre for Research & Technology Hellas (CERTH), PO Box 361, GR-57001 Thessalonica, Greece, Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, 70013 Heraklion, Crete, Greece and Department of Biological Sciences, Bioinformatics Research Laboratory, University of Cyprus, PO Box 20537, CY 1678, Nicosia, Cyprus
| | - Evangelos Pafilis
- Division of Basic Sciences, University of Crete, Medical School, Heraklion 71110, Greece, Institute of Marine Biology, Biotechnology and Aquaculture (IMBBC), Hellenic Centre for Marine Research (HCMR), Heraklion, Greece, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Campus Belval, 7, avenue des Hauts-Fourneaux, L-4362 Esch sur Alzette, Luxembourg, Biological Computation & Process Laboratory (BCPL), Chemical Process & Energy Resources Institute (CPERI), Centre for Research & Technology Hellas (CERTH), PO Box 361, GR-57001 Thessalonica, Greece, Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, 70013 Heraklion, Crete, Greece and Department of Biological Sciences, Bioinformatics Research Laboratory, University of Cyprus, PO Box 20537, CY 1678, Nicosia, Cyprus
| | - Theodosios Theodosiou
- Division of Basic Sciences, University of Crete, Medical School, Heraklion 71110, Greece, Institute of Marine Biology, Biotechnology and Aquaculture (IMBBC), Hellenic Centre for Marine Research (HCMR), Heraklion, Greece, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Campus Belval, 7, avenue des Hauts-Fourneaux, L-4362 Esch sur Alzette, Luxembourg, Biological Computation & Process Laboratory (BCPL), Chemical Process & Energy Resources Institute (CPERI), Centre for Research & Technology Hellas (CERTH), PO Box 361, GR-57001 Thessalonica, Greece, Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, 70013 Heraklion, Crete, Greece and Department of Biological Sciences, Bioinformatics Research Laboratory, University of Cyprus, PO Box 20537, CY 1678, Nicosia, Cyprus
| | - Reinhard Schneider
- Division of Basic Sciences, University of Crete, Medical School, Heraklion 71110, Greece, Institute of Marine Biology, Biotechnology and Aquaculture (IMBBC), Hellenic Centre for Marine Research (HCMR), Heraklion, Greece, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Campus Belval, 7, avenue des Hauts-Fourneaux, L-4362 Esch sur Alzette, Luxembourg, Biological Computation & Process Laboratory (BCPL), Chemical Process & Energy Resources Institute (CPERI), Centre for Research & Technology Hellas (CERTH), PO Box 361, GR-57001 Thessalonica, Greece, Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, 70013 Heraklion, Crete, Greece and Department of Biological Sciences, Bioinformatics Research Laboratory, University of Cyprus, PO Box 20537, CY 1678, Nicosia, Cyprus
| | - Venkata P Satagopam
- Division of Basic Sciences, University of Crete, Medical School, Heraklion 71110, Greece, Institute of Marine Biology, Biotechnology and Aquaculture (IMBBC), Hellenic Centre for Marine Research (HCMR), Heraklion, Greece, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Campus Belval, 7, avenue des Hauts-Fourneaux, L-4362 Esch sur Alzette, Luxembourg, Biological Computation & Process Laboratory (BCPL), Chemical Process & Energy Resources Institute (CPERI), Centre for Research & Technology Hellas (CERTH), PO Box 361, GR-57001 Thessalonica, Greece, Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, 70013 Heraklion, Crete, Greece and Department of Biological Sciences, Bioinformatics Research Laboratory, University of Cyprus, PO Box 20537, CY 1678, Nicosia, Cyprus
| | - Christos A Ouzounis
- Division of Basic Sciences, University of Crete, Medical School, Heraklion 71110, Greece, Institute of Marine Biology, Biotechnology and Aquaculture (IMBBC), Hellenic Centre for Marine Research (HCMR), Heraklion, Greece, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Campus Belval, 7, avenue des Hauts-Fourneaux, L-4362 Esch sur Alzette, Luxembourg, Biological Computation & Process Laboratory (BCPL), Chemical Process & Energy Resources Institute (CPERI), Centre for Research & Technology Hellas (CERTH), PO Box 361, GR-57001 Thessalonica, Greece, Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, 70013 Heraklion, Crete, Greece and Department of Biological Sciences, Bioinformatics Research Laboratory, University of Cyprus, PO Box 20537, CY 1678, Nicosia, Cyprus Division of Basic Sciences, University of Crete, Medical School, Heraklion 71110, Greece, Institute of Marine Biology, Biotechnology and Aquaculture (IMBBC), Hellenic Centre for Marine Research (HCMR), Heraklion, Greece, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Campus Belval, 7, avenue des Hauts-Fourneaux, L-4362 Esch sur Alzette, Luxembourg, Biological Computation & Process Laboratory (BCPL), Chemical Process & Energy Resources Institute (CPERI), Centre for Research & Technology Hellas (CERTH), PO Box 361, GR-57001 Thessalonica, Greece, Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, 70013 Heraklion, Crete, Greece and Department of Biological Sciences, Bioinformatics Research Laboratory, University of Cyprus, PO Box 20537, CY 1678, Nicosia, Cyprus
| | - Aristides G Eliopoulos
- Division of Basic Sciences, University of Crete, Medical School, Heraklion 71110, Greece, Institute of Marine Biology, Biotechnology and Aquaculture (IMBBC), Hellenic Centre for Marine Research (HCMR), Heraklion, Greece, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Campus Belval, 7, avenue des Hauts-Fourneaux, L-4362 Esch sur Alzette, Luxembourg, Biological Computation & Process Laboratory (BCPL), Chemical Process & Energy Resources Institute (CPERI), Centre for Research & Technology Hellas (CERTH), PO Box 361, GR-57001 Thessalonica, Greece, Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, 70013 Heraklion, Crete, Greece and Department of Biological Sciences, Bioinformatics Research Laboratory, University of Cyprus, PO Box 20537, CY 1678, Nicosia, Cyprus Division of Basic Sciences, University of Crete, Medical School, Heraklion 71110, Greece, Institute of Marine Biology, Biotechnology and Aquaculture (IMBBC), Hellenic Centre for Marine Research (HCMR), Heraklion, Greece, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Campus Belval, 7, avenue des Hauts-Fourneaux, L-4362 Esch sur Alzette, Luxembourg, Biological Computation & Process Laboratory (BCPL), Chemical Process & Energy Resources Institute (CPERI), Centre for Research & Technology Hellas (CERTH), PO Box 361, GR-57001 Thessalonica, Greece, Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, 70013 Heraklion, Crete, Greece and Department of Biological Sciences, Bioinformatics Research Laboratory, University of Cyprus, PO Box 20537, CY 1678, Nicosia, Cyprus
| | - Vasilis J Promponas
- Division of Basic Sciences, University of Crete, Medical School, Heraklion 71110, Greece, Institute of Marine Biology, Biotechnology and Aquaculture (IMBBC), Hellenic Centre for Marine Research (HCMR), Heraklion, Greece, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Campus Belval, 7, avenue des Hauts-Fourneaux, L-4362 Esch sur Alzette, Luxembourg, Biological Computation & Process Laboratory (BCPL), Chemical Process & Energy Resources Institute (CPERI), Centre for Research & Technology Hellas (CERTH), PO Box 361, GR-57001 Thessalonica, Greece, Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, 70013 Heraklion, Crete, Greece and Department of Biological Sciences, Bioinformatics Research Laboratory, University of Cyprus, PO Box 20537, CY 1678, Nicosia, Cyprus
| | - Ioannis Iliopoulos
- Division of Basic Sciences, University of Crete, Medical School, Heraklion 71110, Greece, Institute of Marine Biology, Biotechnology and Aquaculture (IMBBC), Hellenic Centre for Marine Research (HCMR), Heraklion, Greece, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Campus Belval, 7, avenue des Hauts-Fourneaux, L-4362 Esch sur Alzette, Luxembourg, Biological Computation & Process Laboratory (BCPL), Chemical Process & Energy Resources Institute (CPERI), Centre for Research & Technology Hellas (CERTH), PO Box 361, GR-57001 Thessalonica, Greece, Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, 70013 Heraklion, Crete, Greece and Department of Biological Sciences, Bioinformatics Research Laboratory, University of Cyprus, PO Box 20537, CY 1678, Nicosia, Cyprus
| |
Collapse
|
39
|
McGuinness B, Craig D, Bullock R, Malouf R, Passmore P, Cochrane Dementia and Cognitive Improvement Group. Statins for the treatment of dementia. Cochrane Database Syst Rev 2014; 2014:CD007514. [PMID: 25004278 PMCID: PMC11112650 DOI: 10.1002/14651858.cd007514.pub3] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND The use of statin therapy in established Alzheimer's disease (AD) or vascular dementia (VaD) is a relatively unexplored area. In AD, β-amyloid protein (Aβ) is deposited in the form of extracellular plaques and previous studies have determined Aβ generation is cholesterol dependent. Hypercholesterolaemia has also been implicated in the pathogenesis of VaD. Due to the role of statins in cholesterol reduction, it is biologically plausible they may be efficacious in the treatment of AD and VaD. OBJECTIVES To assess the clinical efficacy and safety of statins in the treatment of AD and VaD. To evaluate if the efficacy of statins in the treatment of AD and VaD depends on cholesterol level, ApoE genotype or cognitive level. SEARCH METHODS We searched ALOIS, the Specialized Register of the Cochrane Dementia and Cognitive Improvement Group, The Cochrane Library, MEDLINE, EMBASE, PsycINFO, CINAHL and LILACS, as well as many trials registries and grey literature sources (20 January 2014). SELECTION CRITERIA Double-blind, randomised controlled trials of statins given for at least six months in people with a diagnosis of dementia. DATA COLLECTION AND ANALYSIS Two independent authors extracted and assessed data against the inclusion criteria. We pooled data where appropriate and entered them into a meta-analysis. We used standard methodological procedures expected by The Cochrane Collaboration. MAIN RESULTS We identified four studies (1154 participants, age range 50 to 90 years). All participants had a diagnosis of probable or possible AD according to standard criteria and most participants were established on a cholinesterase inhibitor. The primary outcome in all studies was change in Alzheimer's Disease Assessment Scale - cognitive subscale (ADAS-Cog) from baseline. When we pooled data, there was no significant benefit from statin (mean difference -0.26, 95% confidence interval (CI) -1.05 to 0.52, P value = 0.51). All studies provided change in Mini Mental State Examination (MMSE) from baseline. There was no significant benefit from statins in MMSE when we pooled the data (mean difference -0.32, 95% CI -0.71 to 0.06, P value = 0.10). Three studies reported treatment-related adverse effects. When we pooled data, there was no significant difference between statins and placebo (odds ratio 1.09, 95% CI 0.58 to 2.06, P value = 0.78). There was no significant difference in behaviour, global function or activities of daily living in the statin and placebo groups. We assessed risk of bias as low for all studies. We found no studies assessing role of statins in treatment of VaD. AUTHORS' CONCLUSIONS Analyses from the studies available, including two large randomised controlled trials, indicate that statins have no benefit on the primary outcome measures of ADAS-Cog or MMSE.
Collapse
Affiliation(s)
- Bernadette McGuinness
- Belfast Health and Social Care TrustDepartment of Geriatric MedicineLisburn RoadBelfastCo AntrimUK
| | - David Craig
- Craigavon Area HospitalGeriatric MedicineCraigavonNorthern IrelandUK
| | - Roger Bullock
- Kingshill Research Centre, Victoria HospitalOkus RoadSwindonUKSN4 4HZ
| | - Reem Malouf
- University of OxfordNational Perinatal Epidemiology Unit (NPEU)Old Road CampusOxfordUKOX3 7LF
| | - Peter Passmore
- Queen's University BelfastCentre for Public HealthBlock B, ICSB, Grosvenor RoadBelfastNorthern IrelandUKBT12
| | | |
Collapse
|
40
|
Ali-Rahmani F, Schengrund CL, Connor JR. HFE gene variants, iron, and lipids: a novel connection in Alzheimer's disease. Front Pharmacol 2014; 5:165. [PMID: 25071582 PMCID: PMC4086322 DOI: 10.3389/fphar.2014.00165] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Accepted: 06/24/2014] [Indexed: 12/14/2022] Open
Abstract
Iron accumulation and associated oxidative stress in the brain have been consistently found in several neurodegenerative diseases. Multiple genetic studies have been undertaken to try to identify a cause of neurodegenerative diseases but direct connections have been rare. In the iron field, variants in the HFE gene that give rise to a protein involved in cellular iron regulation, are associated with iron accumulation in multiple organs including the brain. There is also substantial epidemiological, genetic, and molecular evidence of disruption of cholesterol homeostasis in several neurodegenerative diseases, in particular Alzheimer's disease (AD). Despite the efforts that have been made to identify factors that can trigger the pathological events associated with neurodegenerative diseases they remain mostly unknown. Because molecular phenotypes such as oxidative stress, synaptic failure, neuronal loss, and cognitive decline, characteristics associated with AD, have been shown to result from disruption of a number of pathways, one can easily argue that the phenotype seen may not arise from a linear sequence of events. Therefore, a multi-targeted approach is needed to understand a complex disorder like AD. This can be achieved only when knowledge about interactions between the different pathways and the potential influence of environmental factors on them becomes available. Toward this end, this review discusses what is known about the roles and interactions of iron and cholesterol in neurodegenerative diseases. It highlights the effects of gene variants of HFE (H63D- and C282Y-HFE) on iron and cholesterol metabolism and how they may contribute to understanding the etiology of complex neurodegenerative diseases.
Collapse
Affiliation(s)
- Fatima Ali-Rahmani
- Departments of Neurosurgery, Neural and Behavioral Sciences and Pediatrics, Center for Aging and Neurodegenerative Diseases, Penn State Hershey Medical CenterHershey, PA, USA
- Departments of Biochemistry and Molecular Biology, The Pennsylvania State University College of MedicineHershey, PA, USA
| | - Cara-Lynne Schengrund
- Departments of Biochemistry and Molecular Biology, The Pennsylvania State University College of MedicineHershey, PA, USA
| | - James R. Connor
- Departments of Neurosurgery, Neural and Behavioral Sciences and Pediatrics, Center for Aging and Neurodegenerative Diseases, Penn State Hershey Medical CenterHershey, PA, USA
| |
Collapse
|
41
|
Cooper SA, Caslake M, Evans J, Hassiotis A, Jahoda A, McConnachie A, Morrison J, Ring H, Starr J, Stiles C, Sullivan F. Toward onset prevention of cognitive decline in adults with Down syndrome (the TOP-COG study): study protocol for a randomized controlled trial. Trials 2014; 15:202. [PMID: 24888381 PMCID: PMC4061534 DOI: 10.1186/1745-6215-15-202] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 05/07/2014] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Early-onset dementia is common in Down syndrome adults, who have trisomy 21. The amyloid precursor protein gene is on chromosome 21, and so is over-expressed in Down syndrome, leading to amyloid β (Aβ) over-production, a major upstream pathway leading to Alzheimer disease (AD). Statins (microsomal 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors), have pleiotropic effects including potentially increasing brain amyloid clearance, making them plausible agents to reduce AD risk. Animal models, human observational studies, and small scale trials support this rationale, however, there are no AD primary prevention trials in Down syndrome adults. In this study we study aim to inform the design of a full-scale primary prevention trial. METHODS/DESIGN TOP-COG is a feasibility and pilot double-blind randomized controlled trial (RCT), with a nested qualitative study, conducted in the general community. About 60 Down syndrome adults, aged ≥50 will be included. The intervention is oral simvastatin 40 mg at night for 12 months, versus placebo. The primary endpoint is recruitment and retention rates. Secondary endpoints are (1) tolerability and safety; (2) detection of the most sensitive neurocognitive instruments; (3) perceptions of Down syndrome adults and caregivers on whether to participate, and assessment experiences; (4) distributions of cognitive decline, adaptive behavior, general health/quality of life, service use, caregiver strain, and sample size implications; (5) whether Aβ42/Aβ40 is a cognitive decline biomarker. We will describe percentages recruited from each source, the number of contacts to achieve this, plus recruitment rate by general population size. We will calculate summary statistics with 90% confidence limits where appropriate, for each study outcome as a whole, by treatment group and in relation to baseline age, cognitive function, cholesterol and other characteristics. Changes over time will be summarized graphically. The sample size for a definitive RCT will be estimated under alternative assumptions. DISCUSSION This study is important, as AD is a major problem for Down syndrome adults, for whom there are currently no effective preventions or treatments. It will also delineate the most suitable assessment instruments for this population. Recruitment of intellectually disabled adults is notoriously difficult, and we shall provide valuable information on this, informing future studies. TRIAL REGISTRATION Current Controlled Trials ISRCTN Register ID: ISRCTN67338640 (17 November 2011).
Collapse
Affiliation(s)
- Sally-Ann Cooper
- Institute of Health and Wellbeing, University of Glasgow, Mental Health and Wellbeing Unit, Gartnavel Royal Hospital, Administrative Building, 1055, Great Western Road, Glasgow G12 0XH, UK
| | - Muriel Caslake
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, McGregor Building, 2nd floor, Western Infirmary, Glasgow G11 6NT, UK
| | - Jonathan Evans
- Institute of Health and Wellbeing, University of Glasgow, Mental Health and Wellbeing Unit, Gartnavel Royal Hospital, Administrative Building, 1055, Great Western Road, Glasgow G12 0XH, UK
| | - Angela Hassiotis
- University College London, Bloomsbury Campus, Charles Bell House, 67-73 Riding House Street, London W1W 7EY, UK
| | - Andrew Jahoda
- Institute of Health and Wellbeing, University of Glasgow, Mental Health and Wellbeing Unit, Gartnavel Royal Hospital, Administrative Building, 1055, Great Western Road, Glasgow G12 0XH, UK
| | - Alex McConnachie
- Robertson Centre for Biostatistics, University of Glasgow, Boyd Orr Building, Glasgow G12 8QQ, UK
| | - Jill Morrison
- Institute of Health and Wellbeing, University of Glasgow, General Practice and Primary Care, 1 Horselethill Road, Glasgow G12 9LX, UK
| | - Howard Ring
- School of Clinical Medicine, University of Cambridge, Douglas House, 18b Trumpington Road, Cambridge CB2 2AH, UK
| | - John Starr
- Alzheimer Scotland Dementia Research Centre, 7 George Square, Edinburgh EH8 9JZ, UK
| | - Ciara Stiles
- Institute of Health and Wellbeing, University of Glasgow, Mental Health and Wellbeing Unit, Gartnavel Royal Hospital, Administrative Building, 1055, Great Western Road, Glasgow G12 0XH, UK
| | - Frank Sullivan
- Gordon F Cheesbrough Research Chair and Director of UTOPIAN, University of Toronto, North York General Hospital, 4001 Leslie Street, Toronto, ON M2K 1E1, Canada
| |
Collapse
|
42
|
Wood WG, Li L, Müller WE, Eckert GP. Cholesterol as a causative factor in Alzheimer's disease: a debatable hypothesis. J Neurochem 2014; 129:559-72. [PMID: 24329875 PMCID: PMC3999290 DOI: 10.1111/jnc.12637] [Citation(s) in RCA: 144] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 11/24/2013] [Accepted: 12/09/2013] [Indexed: 12/17/2022]
Abstract
High serum/plasma cholesterol levels have been suggested as a risk factor for Alzheimer's disease (AD). Some reports, mostly retrospective epidemiological studies, have observed a decreased prevalence of AD in patients taking the cholesterol lowering drugs, statins. The strongest evidence causally linking cholesterol to AD is provided by experimental studies showing that adding/reducing cholesterol alters amyloid precursor protein (APP) and amyloid beta-protein (Ab) levels. However, there are problems with the cholesterol-AD hypothesis. Cholesterol levels in serum/plasma and brain of AD patients do not support cholesterol as a causative factor in AD.Prospective studies on statins and AD have largely failed to show efficacy. Even the experimental data are open to interpretation given that it is well-established that modification of cholesterol levels has effects on multiple proteins, not only amyloid precursor protein and Ab. The purpose of this review, therefore, was to examine the above-mentioned issues, discuss the pros and cons of the cholesterol-AD hypothesis, involvement of other lipids in the mevalonate pathway, and consider that AD may impact cholesterol homeostasis.
Collapse
Affiliation(s)
- W. Gibson Wood
- Geriatric Research, Education and Clinical Center, VAMC, Department of Pharmacology, University of Minnesota School of Medicine, Minneapolis, MN 55455 USA
| | - Ling Li
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455 USA
| | - Walter E. Müller
- Department of Pharmacology, Biocenter Niederursel, Goethe University, Max-von-Laue-St. 9, 60438 Frankfurt, Germany
| | - Gunter P. Eckert
- Department of Pharmacology, Biocenter Niederursel, Goethe University, Max-von-Laue-St. 9, 60438 Frankfurt, Germany
| |
Collapse
|
43
|
Shinohara M, Sato N, Shimamura M, Kurinami H, Hamasaki T, Chatterjee A, Rakugi H, Morishita R. Possible modification of Alzheimer's disease by statins in midlife: interactions with genetic and non-genetic risk factors. Front Aging Neurosci 2014; 6:71. [PMID: 24795626 PMCID: PMC4005936 DOI: 10.3389/fnagi.2014.00071] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2014] [Accepted: 03/30/2014] [Indexed: 12/28/2022] Open
Abstract
The benefits of statins, commonly prescribed for hypercholesterolemia, in treating Alzheimer's disease (AD) have not yet been fully established. A recent randomized clinical trial did not show any therapeutic effects of two statins on cognitive function in AD. Interestingly, however, the results of the Rotterdam study, one of the largest prospective cohort studies, showed reduced risk of AD in statin users. Based on the current understanding of statin actions and AD pathogenesis, it is still worth exploring whether statins can prevent AD when administered decades before the onset of AD or from midlife. This review discusses the possible beneficial effects of statins, drawn from previous clinical observations, pathogenic mechanisms, which include β-amyloid (Aβ) and tau metabolism, genetic and non-genetic risk factors (apolipoprotein E, cholesterol, sex, hypertension, and diabetes), and other clinical features (vascular dysfunction and oxidative and inflammatory stress) of AD. These findings suggest that administration of statins in midlife might prevent AD in late life by modifying genetic and non-genetic risk factors for AD. It should be clarified whether statins inhibit Aβ accumulation, tau pathological features, and brain atrophy in humans. To answer this question, a randomized controlled study using amyloid positron emission tomography (PET), tau-PET, and magnetic resonance imaging would be useful. This clinical evaluation could help us to overcome this devastating disease.
Collapse
Affiliation(s)
- Mitsuru Shinohara
- Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka UniversitySuita, Japan
- Department of Geriatric Medicine, Graduate School of Medicine, Osaka UniversitySuita, Japan
| | - Naoyuki Sato
- Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka UniversitySuita, Japan
- Department of Geriatric Medicine, Graduate School of Medicine, Osaka UniversitySuita, Japan
| | - Munehisa Shimamura
- Division of Vascular Medicine and Epigenetics, Department of Child Development, United Graduate School of Child Development, Osaka University Office for University-Industry CollaborationSuita, Japan
| | - Hitomi Kurinami
- Division of Vascular Medicine and Epigenetics, Department of Child Development, United Graduate School of Child Development, Osaka University Office for University-Industry CollaborationSuita, Japan
| | - Toshimitsu Hamasaki
- Department of Biomedical Statistics, Graduate School of Medicine, Osaka UniversitySuita, Japan
| | - Amarnath Chatterjee
- Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka UniversitySuita, Japan
| | - Hiromi Rakugi
- Department of Geriatric Medicine, Graduate School of Medicine, Osaka UniversitySuita, Japan
| | - Ryuichi Morishita
- Department of Clinical Gene Therapy, Graduate School of Medicine, Osaka UniversitySuita, Japan
| |
Collapse
|
44
|
Rao BS, Gupta KK, Karanam P, Peruri A. Alzheimer disease: An interactome of many diseases. Ann Indian Acad Neurol 2014; 17:48-54. [PMID: 24753659 PMCID: PMC3992769 DOI: 10.4103/0972-2327.128551] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Revised: 07/29/2013] [Accepted: 09/25/2013] [Indexed: 12/14/2022] Open
Abstract
Alzheimer Disease (AD) is an outcome as well as source of many diseases. Alzheimer is linked with many other diseases like Diabetes type 2, cholesterolemia, hypertension and many more. But how each of these diseases affecting other is still unknown to scientific community. Signaling Pathways of one disease is interlinked with other disease. But to what extent healthy brain is affected when any signaling in human body is disturbed is the question that matters. There is a need of Pathway analysis, Protein-Protein interaction (PPI) and the conserved interactome study in AD and linked diseases. It will be helpful in finding the potent drug or vaccine target in conscious manner. In the present research the Protein-Protein interaction of all the proteins involved in Alzheimer Disease is analyzed using ViSANT and osprey tools and pathway analysis further reveals the significant genes/proteins linking AD with other diseases.
Collapse
Affiliation(s)
- Balaji S Rao
- Department of Bioinformatics, NTHRYS Biotech Labs, Hyderabad, Andhra Pradesh, India
| | - Krishna Kant Gupta
- Department of Bioinformatics, NTHRYS Biotech Labs, Hyderabad, Andhra Pradesh, India
| | - Pujitha Karanam
- Department of Bioinformatics, NTHRYS Biotech Labs, Hyderabad, Andhra Pradesh, India
| | - Anusha Peruri
- Department of Bioinformatics, NTHRYS Biotech Labs, Hyderabad, Andhra Pradesh, India
| |
Collapse
|
45
|
Scarpini E, Cogiamanian F. Alzheimer’s disease: from molecular pathogenesis to innovative therapies. Expert Rev Neurother 2014; 3:619-30. [DOI: 10.1586/14737175.3.5.619] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
46
|
Moreira PI, Zhu X, Nunomura A, Smith MA, Perry G. Therapeutic options in Alzheimer’s disease. Expert Rev Neurother 2014; 6:897-910. [PMID: 16784412 DOI: 10.1586/14737175.6.6.897] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Alzheimer's disease (AD) places an enormous burden on individuals, families and society. Consequently, a tremendous effort is being devoted to the development of drugs that prevent or delay neurodegeneration. Current pharmacological treatments are based on the use of acetylcholinesterase inhibitors or memantine, a N-methyl-D-aspartate channel blocker. However, new therapeutic approaches, including those more closely targeted to the pathogenesis of the disease, are being developed. These potentially disease-modifying therapeutics include secretase inhibitors, cholesterol-lowering drugs, amyloid-beta immunotherapy, nonsteroidal anti-inflammatory drugs, hormonal modulation and the use of antioxidants. The possibility that oxidative stress is a primary event in AD indicates that antioxidant-based therapies are perhaps the most promising weapons against this devastating neurodegenerative disorder.
Collapse
Affiliation(s)
- Paula I Moreira
- Case Western Reserve University, Department of Pathology, Cleveland, Ohio 44106, USA.
| | | | | | | | | |
Collapse
|
47
|
Dosunmu R, Wu J, Basha MR, Zawia NH. Environmental and dietary risk factors in Alzheimer’s disease. Expert Rev Neurother 2014; 7:887-900. [PMID: 17610395 DOI: 10.1586/14737175.7.7.887] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease that affects millions in the aging population worldwide and will affect millions more in the next 20 years. Over 90% of all cases are sporadic, with genetics playing a minor role in the etiology of AD. Therefore, it is crucial to investigate the environment and diet as primary risk factors in AD pathology. This review considers epidemiologic case control studies, and in vitro and in vivo research to investigate the potential of environmental exposure to metals, air pollution and pesticides as well as diet as risk factors for AD. In some cases, the role of genetic mutations and environmental risk is discussed. The evidence examined in this review provides a brief overview of the current literature on selected, significant risk factors in promoting amyloid-beta accumulation and aggregation, thus contributing to neurodegeneration.
Collapse
Affiliation(s)
- Remi Dosunmu
- University of Rhode Island, Department of Biomedical & Pharmaceutical Sciences, Kingston, RI 02881, USA.
| | | | | | | |
Collapse
|
48
|
Abstract
Several recent studies demonstrate associations between cardiovascular disease and its risk factors and the incidence of Alzheimer's disease. This review will examine the evidence for these associations and possible pathogenetic pathways. Clinical relevance and implications of these associations will also be discussed.
Collapse
Affiliation(s)
- Caterina Rosano
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| | | |
Collapse
|
49
|
Impact of ApoB-100 expression on cognition and brain pathology in wild-type and hAPPsl mice. Neurobiol Aging 2013; 34:2379-88. [DOI: 10.1016/j.neurobiolaging.2013.04.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Revised: 03/12/2013] [Accepted: 04/03/2013] [Indexed: 11/22/2022]
|
50
|
Fiolaki A, Tsamis KI, Milionis HJ, Kyritsis AP, Kosmidou M, Giannopoulos S. Atherosclerosis, biomarkers of atherosclerosis and Alzheimer's disease. Int J Neurosci 2013; 124:1-11. [DOI: 10.3109/00207454.2013.821988] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|