1
|
Farias HR, Ramos JMO, Griesang CT, Santos L, Junior OVR, Souza DG, Ferreira FS, Somacal S, Martins LAM, de Souza DOG, Moreira JCF, Wyse ATS, Guma FTCR, de Oliveira J. LDL Exposure Disrupts Mitochondrial Function and Dynamics in a Hippocampal Neuronal Cell Line. Mol Neurobiol 2025; 62:6939-6950. [PMID: 39302616 DOI: 10.1007/s12035-024-04476-y] [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: 06/27/2024] [Accepted: 08/30/2024] [Indexed: 09/22/2024]
Abstract
Hypercholesterolemia has been associated with cognitive dysfunction and neurodegenerative diseases. Moreover, this metabolic condition disrupts the blood-brain barrier, allowing low-density lipoprotein (LDL) to enter the central nervous system. Thus, we investigated the effects of LDL exposure on mitochondrial function in a mouse hippocampal neuronal cell line (HT-22). HT-22 cells were exposed to human LDL (50 and 300 μg/mL) for 24 h. After this, intracellular lipid droplet (LD) content, cell viability, cell death, and mitochondrial parameters were assessed. We found that the higher LDL concentration increases LD content compared with control. Both concentrations increased the number of Annexin V-positive cells, indicating apoptosis. Moreover, in mitochondrial parameters, the LDL exposure on hippocampal neuronal cell line leads to a decrease in mitochondrial complexes I and II activities in both concentrations tested and a reduction in Mitotracker™ Red fluorescence and Mitotracker™ Red and Mitotracker™ Green ratio in the higher concentration, indicating mitochondrial impairment. The LDL incubation induces mitochondrial superoxide production and decreases superoxide dismutase activity in the lower concentration in HT-22 cells. Finally, LDL exposure increases the expression of genes associated with mitochondrial fusion (OPA1 and mitofusin 2) in the lower concentration. In conclusion, our findings suggest that LDL exposure induces mitochondrial dysfunction and modulates mitochondrial dynamics in the hippocampal neuronal cells.
Collapse
Affiliation(s)
- Hémelin Resende Farias
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Jessica Marques Obelar Ramos
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Caroline Tainá Griesang
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Lucas Santos
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, Brazil
- Programa de Pós-Graduação em Biologia Celular e Molecular, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Osmar Vieira Ramires Junior
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Debora Guerini Souza
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Fernanda Silva Ferreira
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Sabrina Somacal
- Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Leo Anderson Meira Martins
- Programa de Pós-Graduação em Fisiologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Diogo Onofre Gomes de Souza
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - José Cláudio Fonseca Moreira
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, Brazil
- Programa de Pós-Graduação em Biologia Celular e Molecular, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Angela T S Wyse
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Fátima Theresinha Costa Rodrigues Guma
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Jade de Oliveira
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, Brazil.
| |
Collapse
|
2
|
Tang YY, Lv D. Quantitative proteomic analysis of the protective effect exerted by alliin on ox-LDL-injured HUVECs. Sci Rep 2025; 15:17072. [PMID: 40379781 PMCID: PMC12084529 DOI: 10.1038/s41598-025-01677-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 05/07/2025] [Indexed: 05/19/2025] Open
Abstract
Natural organic sulfides are predominantly found in cruciferous and liliaceous plants. Among these compounds, alliin-an organic sulfide derived from garlic-has garnered significant attention from researchers due to its potential anti-atherosclerotic properties. However, studies specifically investigating the anti-atherosclerotic effects of alliin remain limited. This study aims to elucidate the protective effects of alliin on ox-LDL-injured human umbilical vein endothelial cells (HUVECs) and their underlying mechanisms. Initially, HUVECs were exposed to 80 mg/L oxidized low-density lipoprotein (ox-LDL) for 24 h to establish an ox-LDL injury model. Subsequently, the Cell Counting Kit-8 (CCK8) assay was utilized to assess the effect of alliin on the proliferation of ox-LDL-injured cells at 12, 24 and 48 h, and the levels of total cholesterol (TC) and free cholesterol (Fch) in the HUVECs were measured according to the instructions of TC and Fch kits. Next, quantitative proteomics was then adopted to analyze the differential protein expression in cell samples from the control group (Con), the ox-LDL injury model group (Mod), and the alliin treatment group (Alliin). Among the quantified proteins, a statistical t-test with P < 0.05 was used as a threshold for significance regarding a 1.5-fold change in differential expression. Finally, functional enrichment analysis of the differential proteins in the Alliin/Mod group was performed using Gene Ontology (GO) enrichment and KEGG pathway analysis. Additionally, Western blotting was used to validate the findings. The proteomic analysis identified 6173 identified proteins, of which 5162 were quantifiable. Differential protein analysis revealed that in the Alliin/Con comparison, there were a total of 108 up-regulated proteins and 116 down-regulated proteins; in the Alliin/Mod comparison there were a total of 33 up-regulated proteins and 17 down-regulated proteins; while in the Mod/Con comparison, there were a total of 106 up-regulated proteins and 147 down-regulated proteins. GO enrichment, KEGG pathway analyses and Western blotting verification demonstrated that alliin up-regulates the expression of Low-density lipoprotein receptor (LDLR) (P < 0.05) and apolipoprotein C (ApoC) (P < 0.05) while down-regulating the expression of apolipoprotein B (ApoB) (P < 0.05) to regulate the cholesterol metabolism pathway of the ox-LDL-injured HUVECs. Our findings highlight the importance of cholesterol metabolism in alliin treatment for atherosclerosis. Alliin exerts a protective effect in the ox-LDL-induced HUVEC injury model by modulating the expression of LDLR, ApoC, and ApoB within the cholesterol metabolism pathway. These findings indicate that alliin could potentially serve as a therapeutic agent for the prevention and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Yuan-Yuan Tang
- Department of Pharmacy, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Institute of Material Medica, North Sichuan Medical College, Nanchong, China
| | - De Lv
- Department of Endocrinology, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
3
|
Schoeler T, Pingault JB, Kutalik Z. Combining cross-sectional and longitudinal genomic approaches to identify determinants of cognitive and physical decline. Nat Commun 2025; 16:4524. [PMID: 40374629 DOI: 10.1038/s41467-025-59383-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 04/22/2025] [Indexed: 05/17/2025] Open
Abstract
Large-scale genomic studies focusing on the genetic contribution to human aging have mostly relied on cross-sectional data. With the release of longitudinally curated aging phenotypes by the UK Biobank (UKBB), it is now possible to study aging over time at genome-wide scale. In this work, we evaluated the suitability of competing models of change in realistic simulation settings, performed genome-wide association scans on simulation-validated measures of age-related deweekcline, and followed up with LD-score regression and Mendelian Randomization (MR) analyses. Focusing on global cognitive and physical function, we observed marked differences between baseline function (θ) and accelerated decline (Δ). Both outcomes showed distinct heritability levels (e.g., 31.38%h θ 2 versus 3.15%h Δ 2 for physical function) and different associated loci (e.g., DUSP6 specific to physical Δ). Further, we found little commonalities across the two dimensions of aging-while cognitive decline was largely driven by Alzheimer's disease liability (standardized MR-effect, γ = 0.17), physical decline was mostly impacted by telomere length (γ = -0.05) and bone mineral density (γ = -0.05). Our work highlights the utility of longitudinal genomic efforts to scrutinize age-dependent genetic and environmental effects on physical and cognitive outcomes. Careful modelling and attention to participation characteristics are, however, crucial for valid inference.
Collapse
Affiliation(s)
- Tabea Schoeler
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland.
- Department of Clinical, Educational and Health Psychology, University College London, London, UK.
- Swiss Institute of Bioinformatics, Lausanne, Switzerland.
| | - Jean-Baptiste Pingault
- Department of Clinical, Educational and Health Psychology, University College London, London, UK
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Zoltán Kutalik
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland.
- Swiss Institute of Bioinformatics, Lausanne, Switzerland.
- University Center for Primary Care and Public Health, Lausanne, Switzerland.
| |
Collapse
|
4
|
Zhang Z, Lv L, Guan S, Jiang F, He D, Song H, Sun W, Tian F, Jiang S. Association between serum apolipoprotein B and depression: A cross-sectional and Mendelian randomization analysis study. J Affect Disord 2025; 371:315-323. [PMID: 39579872 DOI: 10.1016/j.jad.2024.11.055] [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: 09/05/2024] [Revised: 11/15/2024] [Accepted: 11/17/2024] [Indexed: 11/25/2024]
Abstract
BACKGROUND Depression is a pervasive mental illness that has a significant impact on public health globally. This study aimed to identify risk factors for depression and elucidate their causal relationships. METHODS Using data from the National Health and Nutrition Examination Survey (NHANES) and Genome-Wide Association Studies (GWAS). Serum ApoB was log-transformed and further divided into 4 groups. Multifactorial logistic regression analysis was used to assess the relationship between serum ApoB and depression. Subgroup analyses and interaction tests were used to observe the stability of the association between them. Smooth curve fitting was used to investigate nonlinear correlations. The causal effect of serum ApoB on depression was assessed using Mendelian randomization (MR) analysis. RESULTS A total of 6531 participated in the study. After adjusting for all covariates, serum ApoB levels were positively associated with depression after adjustment for all covariates (OR = 1.40, 95 % CI = 1.06-1.84; P = 0.0176). Unfortunately, there was no significant causal relationship between serum ApoB and depression (OR = 0.9985,95 % CI = 0.9962-1.0008; P = 0.1923). Sensitivity analysis verified the reliability of the results. CONCLUSIONS Serum ApoB was positively associated with an increased risk of depression, but MR analysis did not show a genetic causal relationship between ApoB and depression. Based on the results of the current study, no indication maintaining high levels of ApoB contributes to the management of depression. LIMITATIONS The main limitation of this study is the inconsistency of the cross-sectional study and the MR population.
Collapse
Affiliation(s)
- Zufa Zhang
- Department of Urology, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, Liaoning, China; Zhongshan Clinical Collage of Dalian University, Dalian 116001, Liaoning, China
| | - Long Lv
- Department of Urology, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, Liaoning, China; Key Laboratory of Microenvironment Regulation and Immunotherapy of Urinary Tumors of Liaoning Province, Dalian 116001, Liaoning, China
| | - Sheng Guan
- Department of Urology, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, Liaoning, China; Key Laboratory of Microenvironment Regulation and Immunotherapy of Urinary Tumors of Liaoning Province, Dalian 116001, Liaoning, China
| | - Fengze Jiang
- Department of Urology, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, Liaoning, China; Key Laboratory of Microenvironment Regulation and Immunotherapy of Urinary Tumors of Liaoning Province, Dalian 116001, Liaoning, China
| | - Danni He
- Department of Urology, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, Liaoning, China; Key Laboratory of Microenvironment Regulation and Immunotherapy of Urinary Tumors of Liaoning Province, Dalian 116001, Liaoning, China
| | - Hongxuan Song
- Department of Urology, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, Liaoning, China; Key Laboratory of Microenvironment Regulation and Immunotherapy of Urinary Tumors of Liaoning Province, Dalian 116001, Liaoning, China
| | - Weibing Sun
- Department of Urology, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, Liaoning, China; Key Laboratory of Microenvironment Regulation and Immunotherapy of Urinary Tumors of Liaoning Province, Dalian 116001, Liaoning, China
| | - Feng Tian
- Department of Urology, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, Liaoning, China; Key Laboratory of Microenvironment Regulation and Immunotherapy of Urinary Tumors of Liaoning Province, Dalian 116001, Liaoning, China.
| | - Sixiong Jiang
- Department of Urology, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, Liaoning, China; Key Laboratory of Microenvironment Regulation and Immunotherapy of Urinary Tumors of Liaoning Province, Dalian 116001, Liaoning, China.
| |
Collapse
|
5
|
Chase BA, Frigerio R, Yucus CJ, Patel S, Maraganore D, Sanders AR, Duan J, Markopoulou K. Lipid trajectories improve risk models for Alzheimer's disease and mild cognitive impairment. J Lipid Res 2025; 66:100714. [PMID: 39586400 PMCID: PMC11731482 DOI: 10.1016/j.jlr.2024.100714] [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: 09/27/2024] [Revised: 11/12/2024] [Accepted: 11/21/2024] [Indexed: 11/27/2024] Open
Abstract
In this retrospective, case-control study, we tested the hypothesis that blood-lipid concentrations during the decade prior to cognitive symptom onset can inform risk prediction for Alzheimer's disease (AD) and stable mild cognitive impairment (MCI). Clinically well-characterized cases were diagnosed using Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition (DSM-IV) criteria; MCI cases had been stable for ≥5 years; and controls were propensity matched to cases at symptom onset (MCI: 116 cases, 435 controls; AD: 215 cases, 483 controls). Participants were grouped based on (i) longitudinal trajectories and (ii) quintile of variability independent of the mean (VIM) for total cholesterol, HDL-C, low-density lipoprotein cholesterol, non-HDL-C, and ln(triglycerides). Risk models evaluated the contributions of lipid trajectory and VIM groups relative to APOE genotype or polygenic risk scores (PRSs) for AD and lipid levels and major lipoprotein confounders: age, lipid-lowering medications, comorbidities, and other longitudinal correlates of blood-lipid concentrations. In models with AD-PRS, higher MCI-risk was associated with the two lower HDL-C trajectories [odds ratios: 3.8(1.3-11.3; P = 0.014), 3.2(1.1-9.3; P = 0.038), relative to the high trajectory], and the lowest VIM quintile of non-HDL-C [odds ratio: 2.2 (1.3-3.8: P = 0.004), relative to quintiles 2-5]. Higher AD-risk was associated with the two lower HDL-C trajectories [odds ratios: 2.8(1.5-5.1; P = 0.001), 3.7 (2.0-7.0; P < 0.001)], and the lowest VIM quintile of total cholesterol [odds ratio: 2.5(1.5-4.0: P < 0.001)]. Inclusion of lipid-trajectory and VIM groups improved risk-model predictive performance independent of APOE and AD or lipid-level PRSs, providing important real-world perspectives on how longitudinal levels and variation of blood-lipid concentrations contribute to risk of cognitive decline.
Collapse
Affiliation(s)
- Bruce A Chase
- Information Technology, Endeavor Health, Skokie, IL, USA; Pritzker School of Medicine, Chicago, USA.
| | - Roberta Frigerio
- Pritzker School of Medicine, Chicago, USA; Research Institute, Endeavor Health, Evanston, IL, USA
| | - Chad J Yucus
- Department of Neurology, Endeavor Health, Evanston, IL, USA
| | - Smita Patel
- Department of Neurology, Endeavor Health, Evanston, IL, USA
| | - Demetrius Maraganore
- Department of Neurology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Alan R Sanders
- Center for Psychiatric Genetics, Endeavor Health Research Institute, Evanston, IL, USA; Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL, USA
| | - Jubao Duan
- Center for Psychiatric Genetics, Endeavor Health Research Institute, Evanston, IL, USA; Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL, USA
| | - Katerina Markopoulou
- Department of Neurology, Endeavor Health, Evanston, IL, USA; Department of Neurology, Pritzker School of Medicine, University of Chicago, Chicago, IL, USA
| |
Collapse
|
6
|
Aumont-Rodrigue G, Picard C, Labonté A, Poirier J. Apolipoprotein B gene expression and regulation in relation to Alzheimer's disease pathophysiology. J Lipid Res 2024; 65:100667. [PMID: 39395793 PMCID: PMC11602985 DOI: 10.1016/j.jlr.2024.100667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 10/04/2024] [Accepted: 10/07/2024] [Indexed: 10/14/2024] Open
Abstract
Apolipoprotein B (APOB), a receptor-binding protein present in cholesterol-rich lipoproteins, has been implicated in Alzheimer's disease (AD). High levels of APOB-containing low-density lipoproteins (LDL) are linked to the pathogenesis of both early-onset familial and late-onset sporadic AD. Rare coding mutations in the APOB gene are associated with familial AD, suggesting a role for APOB-bound lipoproteins in the central nervous system. This research explores APOB gene regulation across the AD spectrum using four cohorts: BRAINEAC (elderly control brains), DBCBB (controls, AD brains), ROSMAP (controls, MCI, AD brains), and ADNI (control, MCI, AD clinical subjects). APOB protein levels, measured via mass spectrometry and ELISA, positively correlated with AD pathology indices and cognition, while APOB mRNA levels showed negative correlations. Brain APOB protein levels are also correlated with cortical Aβ levels. A common coding variant in the APOB gene locus affected its expression but didn't impact AD risk or brain cholesterol concentrations, except for 24-S-hydroxycholesterol. Polymorphisms in the CYP27A1 gene, notably rs4674344, were associated with APOB protein levels. A negative correlation was observed between brain APOB gene expression and AD biomarker levels. CSF APOB correlated with Tau pathology in presymptomatic subjects, while cortical APOB was strongly associated with cortical Aβ deposition in late-stage AD. The study discusses the potential link between blood-brain barrier dysfunction and AD symptoms in relation to APOB neurobiology. Overall, APOB's involvement in lipoprotein metabolism appears to influence AD pathology across different stages of the disease.
Collapse
Affiliation(s)
- Gabriel Aumont-Rodrigue
- Douglas Mental Health University Institute, Neurosciences divison, Montréal, Québec, Canada; Centre for the Studies on Prevention of Alzheimer's Disease, Montréal, Québec, Canada; McGill University, Psychiatry department, Montréal, Québec, Canada
| | - Cynthia Picard
- Douglas Mental Health University Institute, Neurosciences divison, Montréal, Québec, Canada; Centre for the Studies on Prevention of Alzheimer's Disease, Montréal, Québec, Canada
| | - Anne Labonté
- Douglas Mental Health University Institute, Neurosciences divison, Montréal, Québec, Canada; Centre for the Studies on Prevention of Alzheimer's Disease, Montréal, Québec, Canada
| | - Judes Poirier
- Douglas Mental Health University Institute, Neurosciences divison, Montréal, Québec, Canada; Centre for the Studies on Prevention of Alzheimer's Disease, Montréal, Québec, Canada; McGill University, Psychiatry department, Montréal, Québec, Canada.
| |
Collapse
|
7
|
Chase BA, Frigerio R, Yucus CJ, Patel S, Maraganore D, Sanders AR, Duan J, Markopoulou K. Lipid Trajectories Improve Risk Models for Alzheimer's Disease and Mild Cognitive Impairment. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.27.24314494. [PMID: 39399044 PMCID: PMC11469357 DOI: 10.1101/2024.09.27.24314494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
To assess the relationship between lipids and cognitive dysfunction, we retrospectively analyzed blood-lipid levels in clinically well-characterized individuals with stable mild cognitive impairment (MCI) or Alzheimer's disease (AD) over the decade prior to first cognitive symptoms. In this case/control cohort study, AD and MCI cases were diagnosed using DSM-IV criteria; MCI cases had not progressed to dementia for ≥5 years; and controls were propensity matched to cases at age of symptom onset (MCI: 116 cases, 435 controls; AD: 215 cases, 483 controls). Participants were grouped based on longitudinal trajectories and quintile of variability independent of the mean (VIM) for total cholesterol, HDL-C, LDL-C, non-HDL-C and ln(triglycerides). Models for the risk of cognitive dysfunction evaluated trajectory and VIM groups, APOE genotype, polygenic risk scores (PRS) for AD and lipid levels, age, comorbidities, and longitudinal correlates of blood-lipid concentrations. Lower HDL-C trajectories (OR = 3.8, 95% CI = 1.3-11.3) and the lowest VIM quintile of non-HDL-C (OR = 2.2, 95% CI = 1.3-3.0) were associated with higher MCI risk. Lower HDL-C trajectories (OR = 3.0, 95% CI = 1.6-5.7) and the lowest VIM quintile of total cholesterol (OR = 2.4, 95% CI = 1.5-3.9) were associated with higher AD risk. The inclusion of lipid-trajectory and VIM groups improved risk-model predictive performance independent of APOE genotype or PRS for AD and lipid levels. These results provide an important real-world perspective on the influence of lipid metabolism and blood-lipid levels on the development of stable MCI and AD.
Collapse
Affiliation(s)
- Bruce A. Chase
- Information Technology, Endeavor Health, Skokie, IL USA
- Pritzker School of Medicine, Chicago, USA
| | - Roberta Frigerio
- Pritzker School of Medicine, Chicago, USA
- Research Institute, Endeavor Health, Evanston, IL USA
| | - Chad J. Yucus
- Department of Neurology, Endeavor Health, Evanston, IL USA
| | - Smita Patel
- Department of Neurology, Endeavor Health, Evanston, IL USA
| | - Demetrius Maraganore
- Department of Neurology, Tulane University School of Medicine, New Orleans, LA USA
| | - Alan R. Sanders
- Center for Psychiatric Genetics, Endeavor Health Research Institute, Evanston, IL USA
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL USA
| | - Jubao Duan
- Center for Psychiatric Genetics, Endeavor Health Research Institute, Evanston, IL USA
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL USA
| | - Katerina Markopoulou
- Department of Neurology, Endeavor Health, Evanston, IL USA
- Department of Neurology, Pritzker School of Medicine, University of Chicago, Chicago, IL USA
| |
Collapse
|
8
|
Cortes S, Farhat E, Talarico G, Mennigen JA. The dynamic transcriptomic response of the goldfish brain under chronic hypoxia. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2024; 50:101233. [PMID: 38608489 DOI: 10.1016/j.cbd.2024.101233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 03/26/2024] [Accepted: 04/02/2024] [Indexed: 04/14/2024]
Abstract
Oxygen is essential to fuel aerobic metabolism. Some species evolved mechanisms to tolerate periods of severe hypoxia and even anoxia in their environment. Among them, goldfish (Carassius auratus) are unique, in that they do not enter a comatose state under severely hypoxic conditions. There is thus significant interest in the field of comparative physiology to uncover the mechanistic basis underlying hypoxia tolerance in goldfish, with a particular focus on the brain. Taking advantage of the recently published and annotated goldfish genome, we profile the transcriptomic response of the goldfish brain under normoxic (21 kPa oxygen saturation) and, following gradual reduction, constant hypoxic conditions after 1 and 4 weeks (2.1 kPa oxygen saturation). In addition to analyzing differentially expressed protein-coding genes and enriched pathways, we also profile differentially expressed microRNAs (miRs). Using in silico approaches, we identify possible miR-mRNA relationships. Differentially expressed transcripts compared to normoxia were either common to both timepoints of hypoxia exposure (n = 174 mRNAs; n = 6 miRs), or exclusive to 1-week (n = 441 mRNAs; n = 23 miRs) or 4-week hypoxia exposure (n = 491 mRNAs; n = 34 miRs). Under chronic hypoxia, an increasing number of transcripts, including those of paralogous genes, was downregulated over time, suggesting a decrease in transcription. GO-terms related to the vascular system, oxidative stress, stress signalling, oxidoreductase activity, nucleotide- and intermediary metabolism, and mRNA posttranscriptional regulation were found to be enriched under chronic hypoxia. Known 'hypoxamiRs', such as miR-210-3p/5p, and miRs such as miR-29b-3p likely contribute to posttranscriptional regulation of these pathways under chronic hypoxia in the goldfish brain.
Collapse
Affiliation(s)
- S Cortes
- Department of Biology, University of Ottawa, K1N6N5 20 Marie Curie, Ottawa, ON, Canada; Laboratorio de Oncogenómica, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City 14610, Mexico
| | - E Farhat
- Department of Biology, University of Ottawa, K1N6N5 20 Marie Curie, Ottawa, ON, Canada; Department of Biosciences, Faculty of Mathematics and Natural Sciences, University of Oslo, 0371 Oslo, Norway
| | - Ggm Talarico
- Department of Biology, University of Ottawa, K1N6N5 20 Marie Curie, Ottawa, ON, Canada
| | - J A Mennigen
- Department of Biology, University of Ottawa, K1N6N5 20 Marie Curie, Ottawa, ON, Canada.
| |
Collapse
|
9
|
Martin L, Boutwell BB, Messerlian C, Adams CD. Mendelian randomization reveals apolipoprotein B shortens healthspan and possibly increases risk for Alzheimer's disease. Commun Biol 2024; 7:230. [PMID: 38402277 PMCID: PMC10894226 DOI: 10.1038/s42003-024-05887-2] [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: 12/29/2022] [Accepted: 02/05/2024] [Indexed: 02/26/2024] Open
Abstract
Apolipoprotein B-100 (APOB) is a component of fat- and cholesterol-transporting molecules in the bloodstream. It is the main lipoprotein in low-density lipoprotein cholesterol (LDL) and has been implicated in conditions that end healthspan (the interval between birth and onset of chronic disease). However, APOB's direct relationship with healthspan remains uncertain. With Mendelian randomization, we show that higher levels of APOB and LDL shorten healthspan in humans. Multivariable Mendelian randomization of APOB and LDL on healthspan suggests that the predominant trait accounting for the relationship is APOB. In addition, we provide preliminary evidence that APOB increases risk for Alzheimer's disease, a condition that ends healthspan. If these relationships are causal, they suggest that interventions to improve healthspan in aging populations could include strategies targeting APOB. Ultimately, given that more than 44 million people currently suffer from Alzheimer's disease worldwide, such interventions are needed.
Collapse
Affiliation(s)
- Leah Martin
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Brian B Boutwell
- School of Applied Sciences, University of Mississippi, University, Jackson, MS, USA
- John D. Bower School of Population Health, University of Mississippi Medical Center, Jackson, MS, USA
| | - Carmen Messerlian
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Department of Obstetrics and Gynecology, Massachusetts General Hospital Fertility Center, Boston, MA, USA
- Department of Environmental Health, Harvard T. H. Chan School of Public Heath, Boston, MA, USA
| | - Charleen D Adams
- Department of Environmental Health, Harvard T. H. Chan School of Public Heath, Boston, MA, USA.
| |
Collapse
|
10
|
de Oliveira J, Moreira ELG, de Bem AF. Beyond cardiovascular risk: Implications of Familial hypercholesterolemia on cognition and brain function. Ageing Res Rev 2024; 93:102149. [PMID: 38056504 DOI: 10.1016/j.arr.2023.102149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 11/20/2023] [Accepted: 11/30/2023] [Indexed: 12/08/2023]
Abstract
Familial hypercholesterolemia (FH) is a metabolic condition caused mainly by a mutation in the low-density lipoprotein (LDL) receptor gene (LDLR), which is highly prevalent in the population. Besides being an important causative factor of cardiovascular diseases, FH has been considered an early risk factor for Alzheimer's disease. Cognitive and emotional behavioral impairments in LDL receptor knockout (LDLr-/-) mice are associated with neuroinflammation, blood-brain barrier dysfunction, impaired neurogenesis, brain oxidative stress, and mitochondrial dysfunction. Notably, today, LDLr-/- mice, a widely used animal model for studying cardiovascular diseases and atherosclerosis, are also considered an interesting tool for studying dementia. Here, we reviewed the main findings in LDLr-/- mice regarding the relationship between FH and brain dysfunctions and dementia development.
Collapse
Affiliation(s)
- Jade de Oliveira
- Laboratory of investigation on metabolic disorders and neurodegenerative diseases, Department of Biochemistry, Federal University of Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil.
| | - Eduardo Luiz Gasnhar Moreira
- Neuroscience Coworking Lab, Department of Physiological Sciences, Federal University of Santa Catarina, Florianópolis, SC 88040-900, Brazil.
| | - Andreza Fabro de Bem
- Laboratory of Bioenergetics and Metabolism, Department of Physiological Sciences, University of Brasilia, Brasília, Federal District, DF 70910-900, Brazil; Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden; Brazilian National Institute of Science and Technology on Neuroimmunomodulation, Oswaldo Cruz Foundation, Rio de Janeiro, RJ 21040360, Brazil.
| |
Collapse
|
11
|
Kakava S, Schlumpf E, Panteloglou G, Tellenbach F, von Eckardstein A, Robert J. Brain Endothelial Cells in Contrary to the Aortic Do Not Transport but Degrade Low-Density Lipoproteins via Both LDLR and ALK1. Cells 2022; 11:cells11193044. [PMID: 36231005 PMCID: PMC9564369 DOI: 10.3390/cells11193044] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/23/2022] [Accepted: 09/26/2022] [Indexed: 11/16/2022] Open
Abstract
The transport of low-density lipoprotein (LDL) through the endothelium is a key step in the development of atherosclerosis, but it is notorious that phenotypic differences exist between endothelial cells originating from different vascular beds. Endothelial cells forming the blood–brain barrier restrict paracellular and transcellular passage of plasma proteins. Here, we systematically compared brain versus aortic endothelial cells towards their interaction with LDL and the role of proteins known to regulate the uptake of LDL by endothelial cells. Both brain endothelial cells and aortic endothelial cells bind and internalize LDL. However, whereas aortic endothelial cells degrade very small amounts of LDL and transcytose the majority, brain endothelial cells degrade but do not transport LDL. Using RNA interference (siRNA), we found that the LDLR–clathrin pathway leads to LDL degradation in either endothelial cell type. Both loss- and gain-of-function experiments showed that ALK1, which promotes transcellular LDL transport in aortic endothelial cells, also limits LDL degradation in brain endothelial cells. SR-BI and caveolin-1, which promote LDL uptake and transport into aortic endothelial cells, limit neither binding nor association of LDL to brain endothelial cells. Together, these results indicate distinct LDL trafficking by brain microvascular endothelial cells and aortic endothelial cells.
Collapse
Affiliation(s)
- Sofia Kakava
- Institute of Clinical Chemistry, University Hospital of Zurich, 8952 Schlieren, Switzerland
- Bio Medicine Program, Life Science Zurich Graduate School, University of Zurich, 8000 Zurich, Switzerland
| | - Eveline Schlumpf
- Institute of Clinical Chemistry, University Hospital of Zurich, 8952 Schlieren, Switzerland
| | - Grigorios Panteloglou
- Institute of Clinical Chemistry, University Hospital of Zurich, 8952 Schlieren, Switzerland
| | - Flavia Tellenbach
- Institute of Clinical Chemistry, University Hospital of Zurich, 8952 Schlieren, Switzerland
| | - Arnold von Eckardstein
- Institute of Clinical Chemistry, University Hospital of Zurich, 8952 Schlieren, Switzerland
- Bio Medicine Program, Life Science Zurich Graduate School, University of Zurich, 8000 Zurich, Switzerland
| | - Jerome Robert
- Institute of Clinical Chemistry, University Hospital of Zurich, 8952 Schlieren, Switzerland
- Correspondence: or
| |
Collapse
|
12
|
Huang SY, Yang YX, Zhang YR, Kuo K, Li HQ, Shen XN, Chen SD, Chen KL, Dong Q, Tan L, Yu JT. Investigating Causal Relations Between Circulating Metabolites and Alzheimer's Disease: A Mendelian Randomization Study. J Alzheimers Dis 2022; 87:463-477. [PMID: 35275550 DOI: 10.3233/jad-220050] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
BACKGROUND Metabolomics is a promising approach that can be used to understand pathophysiological pathways of Alzheimer's disease (AD). However, the causal relationships between metabolism and AD are poorly understood. OBJECTIVE We aimed to investigate the causal association between circulating metabolites and risk of AD through two-sample Mendelian randomization (MR) approach. METHODS Genetic associations with 123 circulating metabolic traits were utilized as exposures. Summary statistics data from International Genomics of Alzheimer's Project was used in primary analysis, including 21,982 AD cases and 41,944 controls. Validation was performed using family history of AD data from UK Biobank (27,696 cases of maternal AD, 14,338 cases of paternal AD, and 272,244 controls). We utilized inverse-variance weighted method as primary method. RESULTS We found significantly increased risks of developing AD per standard deviation increase in the levels of circulating ApoB (odd ratio[OR] = 3.18; 95% confidence interval[CI]: 1.52-6.66, p = 0.0022), glycoprotein acetyls (OR = 1.21; 95% CI: 1.05-1.39, p = 0.0093), total cholesterol (OR = 2.73; 95% CI: 1.41-5.30, p = 0.0030), and low-density lipoprotein (LDL) cholesterol (OR = 2.34; 95% CI: 1.53-3.57, p = 0.0001). Whereas glutamine (OR = 0.81; 95% CI: 0.71-0.92, p = 0.0011) were significantly associated with lower risk of AD. We also detected causal effects of several different composition of LDL fractions on increased AD risk, which has been verified in validation. However, we found no association between circulating high-density lipoprotein cholesterol and AD. CONCLUSION Our findings suggest causal effects of circulating glycoprotein acetyls, ApoB, LDL cholesterol, and serum total cholesterol on higher risk of AD, whereas glutamine showed the protective effect.
Collapse
Affiliation(s)
- Shu-Yi Huang
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, China
| | - Yu-Xiang Yang
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, China
| | - Ya-Ru Zhang
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, China
| | - Kevin Kuo
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, China
| | - Hong-Qi Li
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, China
| | - Xue-Ning Shen
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, China
| | - Shi-Dong Chen
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, China
| | - Ke-Liang Chen
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, China
| | - Qiang Dong
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, China
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, China
| |
Collapse
|
13
|
Picard C, Nilsson N, Labonté A, Auld D, Rosa-Neto P, Ashton NJ, Zetterberg H, Blennow K, Breitner JCB, Villeneuve S, Poirier J. Apolipoprotein B is a novel marker for early tau pathology in Alzheimer's disease. Alzheimers Dement 2021; 18:875-887. [PMID: 34590423 PMCID: PMC9293308 DOI: 10.1002/alz.12442] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 06/30/2021] [Accepted: 07/05/2021] [Indexed: 01/08/2023]
Abstract
INTRODUCTION We examine the role of brain apolipoprotein B (apoB) as a putative marker of early tau pathology and cognitive decline. METHODS Cerebrospinal fluid (CSF) samples from cognitively normal and Alzheimer's disease (AD) participants were collected to measure protein levels of apoB and AD biomarkers amyloid beta (Aβ), t-tau and p-tau, as well as synaptic markers GAP43, SYNAPTOTAGMIN-1, synaptosome associated protein 25 (SNAP-25), and NEUROGRANIN. CSF apoB levels were contrasted with positron emission tomography (PET) scan measures of Aβ (18F-NAV4694) and Tau (flortaucipir) along with cognitive assessment alterations over 6 to 8 years. RESULTS CSF apoB levels were elevated in AD participants and correlated with t-tau, p-tau, and the four synaptic markers in pre-symptomatic individuals. In the latter, CSF apoB levels correlated with PET flortaucipir-binding in entorhinal, parahippocampal, and fusiform regions. Baseline CSF apoB levels were associated with longitudinal visuospatial cognitive decline. DISCUSSION CSF apoB markedly associates with early tau dysregulation in asymptomatic subjects and identifies at-risk individuals predisposed to develop visuospatial cognitive decline over time.
Collapse
Affiliation(s)
- Cynthia Picard
- Douglas Mental Health University Institute, Montréal, Québec, Canada.,Centre for the Studies in the Prevention of Alzheimer's Disease, Montréal, Québec, Canada
| | - Nathalie Nilsson
- Douglas Mental Health University Institute, Montréal, Québec, Canada.,Centre for the Studies in the Prevention of Alzheimer's Disease, Montréal, Québec, Canada.,McGill University, Montréal, Québec, Canada
| | - Anne Labonté
- Douglas Mental Health University Institute, Montréal, Québec, Canada.,Centre for the Studies in the Prevention of Alzheimer's Disease, Montréal, Québec, Canada
| | | | - Pedro Rosa-Neto
- Douglas Mental Health University Institute, Montréal, Québec, Canada.,Centre for the Studies in the Prevention of Alzheimer's Disease, Montréal, Québec, Canada.,McGill University, Montréal, Québec, Canada
| | -
- Douglas Mental Health University Institute, Montréal, Québec, Canada
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK.,UK Dementia Research Institute at UCL, London, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - John C B Breitner
- Douglas Mental Health University Institute, Montréal, Québec, Canada.,Centre for the Studies in the Prevention of Alzheimer's Disease, Montréal, Québec, Canada.,McGill University, Montréal, Québec, Canada
| | - Sylvia Villeneuve
- Douglas Mental Health University Institute, Montréal, Québec, Canada.,Centre for the Studies in the Prevention of Alzheimer's Disease, Montréal, Québec, Canada.,McGill University, Montréal, Québec, Canada
| | - Judes Poirier
- Douglas Mental Health University Institute, Montréal, Québec, Canada.,Centre for the Studies in the Prevention of Alzheimer's Disease, Montréal, Québec, Canada.,McGill University, Montréal, Québec, Canada
| | -
- Douglas Mental Health University Institute, Montréal, Québec, Canada
| |
Collapse
|
14
|
Kalli ΕG. The Effect of Nutrients on Alzheimer’s Disease Biomarkers: A Metabolomic Approach. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1339:301-308. [DOI: 10.1007/978-3-030-78787-5_35] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
15
|
Tóth ME, Dukay B, Hoyk Z, Sántha M. Cerebrovascular Changes and Neurodegeneration Related to Hyperlipidemia: Characteristics of the Human ApoB-100 Transgenic Mice. Curr Pharm Des 2020; 26:1486-1494. [PMID: 32067608 PMCID: PMC7403644 DOI: 10.2174/1381612826666200218101818] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 11/27/2019] [Indexed: 01/07/2023]
Abstract
Serum lipid levels are closely related to the structure and function of blood vessels. Chronic hyperlipidemia may lead to damage in both the cardio- and the cerebrovascular systems. Vascular dysfunctions, including impairments of the blood-brain barrier, are known to be associated with neurodegenerative diseases. A growing number of evidence suggests that cardiovascular risk factors, such as hyperlipidemia, may increase the likelihood of developing dementia. Due to differences in lipoprotein metabolism, wild-type mice are protected against diet-induced hypercholesterolemia, and their serum lipid profile is different from that observed in humans. Therefore, several transgenic mouse models have been established to study the role of different apolipoproteins and their receptors in lipid metabolism, as well as the complications related to pathological lipoprotein levels. This mini-review focused on a transgenic mouse model overexpressing an apolipoprotein, the human ApoB-100. We discussed literature data and current advancements on the understanding of ApoB-100 induced cardio- and cerebrovascular lesions in order to demonstrate the involvement of this type of apolipoprotein in a wide range of pathologies, and a link between hyperlipidemia and neurodegeneration.
Collapse
Affiliation(s)
- Melinda E Tóth
- Institute of Biochemistry, Biological Research Centre, Sezeged, Hungary
| | - Brigitta Dukay
- Institute of Biochemistry, Biological Research Centre, Sezeged, Hungary.,Doctoral School in Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Zsófia Hoyk
- Institute of Biophysics, Biological Research Centre, H-6726 Szeged, Temesvári krt. 62., Hungary
| | - Miklós Sántha
- Institute of Biochemistry, Biological Research Centre, Sezeged, Hungary
| |
Collapse
|
16
|
Diniz Pereira J, Gomes Fraga V, Morais Santos AL, Carvalho MDG, Caramelli P, Braga Gomes K. Alzheimer's disease and type 2 diabetes mellitus: A systematic review of proteomic studies. J Neurochem 2020; 156:753-776. [PMID: 32909269 DOI: 10.1111/jnc.15166] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 07/15/2020] [Accepted: 08/25/2020] [Indexed: 12/16/2022]
Abstract
Similar to dementia, the risk for developing type 2 diabetes mellitus (T2DM) increases with age, and T2DM also increases the risk for dementia, particularly Alzheimer's disease (AD). Although T2DM is primarily a peripheral disorder and AD is a central nervous system disease, both share some common features as they are chronic and complex diseases, and both show involvement of oxidative stress and inflammation in their progression. These characteristics suggest that T2DM may be associated with AD, which gave rise to a new term, type 3 diabetes (T3DM). In this study, we searched for matching peripheral proteomic biomarkers of AD and T2DM based in a systematic review of the available literature. We identified 17 common biomarkers that were differentially expressed in both patients with AD or T2DM when compared with healthy controls. These biomarkers could provide a useful workflow for screening T2DM patients at risk to develop AD.
Collapse
Affiliation(s)
- Jessica Diniz Pereira
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Vanessa Gomes Fraga
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Anna Luiza Morais Santos
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Maria das Graças Carvalho
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Paulo Caramelli
- Departamento de Clínica Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Karina Braga Gomes
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
17
|
Neddens J, Daurer M, Flunkert S, Beutl K, Loeffler T, Walker L, Attems J, Hutter-Paier B. Correlation of pyroglutamate amyloid β and ptau Ser202/Thr205 levels in Alzheimer's disease and related murine models. PLoS One 2020; 15:e0235543. [PMID: 32645028 PMCID: PMC7347153 DOI: 10.1371/journal.pone.0235543] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 06/17/2020] [Indexed: 01/14/2023] Open
Abstract
Senile plaques frequently contain Aβ-pE(3), a N-terminally truncated Aβ species that is more closely linked to AD compared to other Aβ species. Tau protein is highly phosphorylated at several residues in AD, and specifically phosphorylation at Ser202/Thr205 is known to be increased in AD. Several studies suggest that formation of plaques and tau phosphorylation might be linked to each other. To evaluate if Aβ-pE(3) and ptau Ser202/Thr205 levels correlate in human and transgenic AD mouse models, we analyzed human cortical and hippocampal brain tissue of different Braak stages as well as murine brain tissue of two transgenic mouse models for levels of Aβ-pE(3) and ptau Ser202/Thr205 and correlated the data. Our results show that Aβ-pE(3) formation is increased at early Braak stages while ptau Ser202/Thr205 mostly increases at later stages. Further analyses revealed strongest correlations between the two pathologies in the temporal, frontal, cingulate, and occipital cortex, however correlation in the hippocampus was weaker. Evaluation of murine transgenic brain tissue demonstrated a slow but steady increase of Aβ-pE(3) from 6 to 12 months of age in the cortex and hippocampus of APPSL mice, and a very early and strong Aβ-pE(3) increase in 5xFAD mice. ptau Ser202/Thr205 levels increased at the age of 9 months in APPSL mice and at 6 months in 5xFAD mice. Our results show that Aβ-pE(3) and ptau Ser202/Thr205 levels strongly correlate in human as well as murine tissues, suggesting that tau phosphorylation might be amplified by Aβ-pE(3).
Collapse
Affiliation(s)
| | | | | | - Kerstin Beutl
- QPS Austria GmbH, Grambach, Austria
- FH Joanneum Graz, Graz, Austria
| | | | - Lauren Walker
- Translational and Clinical Research Institute and Newcastle University Institute for Ageing, Campus for Ageing and Vitality, Newcastle upon Tyne, United Kingdom
| | - Johannes Attems
- Translational and Clinical Research Institute and Newcastle University Institute for Ageing, Campus for Ageing and Vitality, Newcastle upon Tyne, United Kingdom
| | | |
Collapse
|
18
|
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
|
19
|
Engel DF, Grzyb AN, de Oliveira J, Pötzsch A, Walker TL, Brocardo PS, Kempermann G, de Bem AF. Impaired adult hippocampal neurogenesis in a mouse model of familial hypercholesterolemia: A role for the LDL receptor and cholesterol metabolism in adult neural precursor cells. Mol Metab 2019; 30:1-15. [PMID: 31767163 PMCID: PMC6812372 DOI: 10.1016/j.molmet.2019.09.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/02/2019] [Accepted: 09/04/2019] [Indexed: 02/07/2023] Open
Abstract
Objective In familial hypercholesterolemia (FH), mutations in the low-density lipoprotein (LDL) receptor (LDLr) gene result in increased plasma LDL cholesterol. Clinical and preclinical studies have revealed an association between FH and hippocampus-related memory and mood impairment. We here asked whether hippocampal pathology in FH might be a consequence of compromised adult hippocampal neurogenesis. Methods We evaluated hippocampus-dependent behavior and neurogenesis in adult C57BL/6JRj and LDLr−/− mice. We investigated the effects of elevated cholesterol and the function of LDLr in neural precursor cells (NPC) isolated from adult C57BL/6JRj mice in vitro. Results Behavioral tests revealed that adult LDLr−/− mice showed reduced performance in a dentate gyrus (DG)-dependent metric change task. This phenotype was accompanied by a reduction in cell proliferation and adult neurogenesis in the DG of LDLr−/− mice, suggesting a potential direct impact of LDLr mutation on NPC. Exposure of NPC to LDL as well as LDLr gene knockdown reduced proliferation and disrupted transcriptional activity of genes involved in endogenous cholesterol synthesis and metabolism. The LDL treatment also induced an increase in intracellular lipid storage. Functional analysis of differentially expressed genes revealed parallel modulation of distinct regulatory networks upon LDL treatment and LDLr knockdown. Conclusions Together, these results suggest that high LDL levels and a loss of LDLr function, which are characteristic to individuals with FH, might contribute to a disease-related impairment in adult hippocampal neurogenesis and, consequently, cognitive functions. The LDLr −/− mice show impaired hippocampal related behaviour and adult neurogenesis. In vitro exposure of NPC to LDL and LDLr knock-down reduces cell proliferation. LDL exposure induces lipid storage in NPC. In vitro LDL and LDLr knock-down in NPC modulates distinct regulatory networks.
Collapse
Affiliation(s)
- Daiane F Engel
- Department of Biochemistry, Federal University of Santa Catarina, Florianópolis, Brazil; German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany.
| | - Anna N Grzyb
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany; CRTD - Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Jade de Oliveira
- Department of Biochemistry, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Alexandra Pötzsch
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany; CRTD - Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Tara L Walker
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany; CRTD - Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Patricia S Brocardo
- Department of Morphological Sciences, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Gerd Kempermann
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany; CRTD - Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Andreza F de Bem
- Department of Biochemistry, Federal University of Santa Catarina, Florianópolis, Brazil; Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, Brazil.
| |
Collapse
|
20
|
Affiliation(s)
- Makoto Ishii
- Feil Family Brain and Mind Research Institute, Department of Neurology, Weill Cornell Medicine, New York, New York
| |
Collapse
|
21
|
Hoyk Z, Tóth ME, Lénárt N, Nagy D, Dukay B, Csefová A, Zvara Á, Seprényi G, Kincses A, Walter FR, Veszelka S, Vígh J, Barabási B, Harazin A, Kittel Á, Puskás LG, Penke B, Vígh L, Deli MA, Sántha M. Cerebrovascular Pathology in Hypertriglyceridemic APOB-100 Transgenic Mice. Front Cell Neurosci 2018; 12:380. [PMID: 30410436 PMCID: PMC6209654 DOI: 10.3389/fncel.2018.00380] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 10/04/2018] [Indexed: 01/08/2023] Open
Abstract
Hypertriglyceridemia is not only a serious risk factor in the development of cardiovascular diseases, but it is linked to neurodegeneration, too. Previously, we generated transgenic mice overexpressing the human APOB-100 protein, a mouse model of human atherosclerosis. In this model we observed high plasma levels of triglycerides, oxidative stress, tau hyperphosphorylation, synaptic dysfunction, cognitive impairment, increased neural apoptosis and neurodegeneration. Neurovascular dysfunction is recognized as a key factor in the development of neurodegenerative diseases, but the cellular and molecular events linking cerebrovascular pathology and neurodegeneration are not fully understood. Our aim was to study cerebrovascular changes in APOB-100 transgenic mice. We described the kinetics of the development of chronic hypertriglyceridemia in the transgenic animals. Increased blood-brain barrier permeability was found in the hippocampus of APOB-100 transgenic mice which was accompanied by structural changes. Using transmission electron microscopy, we detected changes in the brain capillary endothelial tight junction structure and edematous swelling of astrocyte endfeet. In brain microvessels isolated from APOB-100 transgenic animals increased Lox-1, Aqp4, and decreased Meox-2, Mfsd2a, Abcb1a, Lrp2, Glut-1, Nos2, Nos3, Vim, and in transgenic brains reduced Cdh2 and Gfap-σ gene expressions were measured using quantitative real-time PCR. We confirmed the decreased P-glycoprotein (ABCB1) and vimentin expression related to the neurovascular unit by immunostaining in transgenic brain sections using confocal microscopy. We conclude that in chronic hypertriglyceridemic APOB-100 transgenic mice both functional and morphological cerebrovascular pathology can be observed, and this animal model could be a useful tool to study the link between cerebrovascular pathology and neurodegeneration.
Collapse
Affiliation(s)
- Zsófia Hoyk
- Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Melinda E Tóth
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Nikolett Lénárt
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Dóra Nagy
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Brigitta Dukay
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Alexandra Csefová
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Ágnes Zvara
- Laboratory of Functional Genomics, Core Facilities, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - György Seprényi
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - András Kincses
- Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Fruzsina R Walter
- Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Szilvia Veszelka
- Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Judit Vígh
- Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Beáta Barabási
- Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - András Harazin
- Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Ágnes Kittel
- Laboratory of Molecular Pharmacology, Department of Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - László G Puskás
- Laboratory of Functional Genomics, Core Facilities, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Botond Penke
- Department of Medical Chemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - László Vígh
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Mária A Deli
- Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Miklós Sántha
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| |
Collapse
|
22
|
Martiskainen H, Takalo M, Solomon A, Stančáková A, Marttinen M, Natunen T, Haapasalo A, Herukka SK, Kuusisto J, Soininen H, Kivipelto M, Laakso M, Hiltunen M. Decreased plasma C-reactive protein levels in APOE ε4 allele carriers. Ann Clin Transl Neurol 2018; 5:1229-1240. [PMID: 30349858 PMCID: PMC6186931 DOI: 10.1002/acn3.639] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 08/07/2018] [Accepted: 08/10/2018] [Indexed: 02/06/2023] Open
Abstract
Objective Apolipoprotein E (APOE) ε4 allele is a well‐established risk factor in Alzheimer's disease (AD). Here, we assessed the effects of APOE polymorphism on cardiovascular, metabolic, and inflammation‐related parameters in population‐based cohorts. Methods Association of cardiovascular, metabolic, and inflammation‐related parameters with the APOE polymorphism in a large Finnish Metabolic Syndrome in Men (METSIM) cohort and Finnish Geriatric Intervention study to prevent cognitive impairment and disability (FINGER) were investigated. Brain‐specific effects were addressed in postmortem brain samples. Results Individuals carrying the APOE ε4 allele displayed significantly elevated serum/plasma LDL cholesterol and apolipoprotein B levels. APOE ε3ε4 and ε4ε4 significantly associated with lower levels of plasma high‐sensitivity C‐reactive protein (hs‐CRP). Plasma amyloid‐β 42 (Aβ42) and reduced hs‐CRP levels showed an association independently of the APOE status. Interpretation These data suggest that the APOE ε4 allele associates with lower levels of hs‐CRP in individuals without dementia. Moreover, Aβ42 may encompass anti‐inflammatory effects reflected by reduced hs‐CRP levels.
Collapse
Affiliation(s)
- Henna Martiskainen
- Institute of Clinical Medicine Internal Medicine University of Eastern Finland P.O. Box 1627 Kuopio 70211 Finland
| | - Mari Takalo
- Institute of Biomedicine University of Eastern Finland Yliopistonranta 1 E, P.O. Box 1627 Kuopio 70211 Finland
| | - Alina Solomon
- Department of Neurology Institute of Clinical Medicine University of Eastern Finland P.O. Box 1627 Kuopio 70211 Finland.,Division of Clinical Geriatrics Center for Alzheimer Research NVS, Karolinska Institutet Novum 5th floor Huddinge 14157 Sweden
| | - Alena Stančáková
- Institute of Clinical Medicine Internal Medicine University of Eastern Finland P.O. Box 1627 Kuopio 70211 Finland
| | - Mikael Marttinen
- Institute of Biomedicine University of Eastern Finland Yliopistonranta 1 E, P.O. Box 1627 Kuopio 70211 Finland
| | - Teemu Natunen
- Institute of Biomedicine University of Eastern Finland Yliopistonranta 1 E, P.O. Box 1627 Kuopio 70211 Finland
| | - Annakaisa Haapasalo
- A.I Virtanen Institute for Molecular Sciences University of Eastern Finland Neulaniementie 2 Kuopio 70211 Finland
| | - Sanna-Kaisa Herukka
- Department of Neurology Institute of Clinical Medicine University of Eastern Finland P.O. Box 1627 Kuopio 70211 Finland.,Neurocenter Neurology Kuopio University Hospital Kuopio Finland
| | - Johanna Kuusisto
- Institute of Clinical Medicine Internal Medicine University of Eastern Finland P.O. Box 1627 Kuopio 70211 Finland.,Kuopio University Hospital Kuopio Finland
| | - Hilkka Soininen
- Department of Neurology Institute of Clinical Medicine University of Eastern Finland P.O. Box 1627 Kuopio 70211 Finland.,Neurocenter Neurology Kuopio University Hospital Kuopio Finland
| | - Miia Kivipelto
- Department of Neurology Institute of Clinical Medicine University of Eastern Finland P.O. Box 1627 Kuopio 70211 Finland.,Division of Clinical Geriatrics Center for Alzheimer Research NVS, Karolinska Institutet Novum 5th floor Huddinge 14157 Sweden.,Department of Public Health Solutions Public Health Promotion Unit National Institute for Health and Welfare PO Box 30 Helsinki 00271 Finland
| | - Markku Laakso
- Institute of Clinical Medicine Internal Medicine University of Eastern Finland P.O. Box 1627 Kuopio 70211 Finland.,Kuopio University Hospital Kuopio Finland
| | - Mikko Hiltunen
- Institute of Biomedicine University of Eastern Finland Yliopistonranta 1 E, P.O. Box 1627 Kuopio 70211 Finland
| |
Collapse
|
23
|
Latta-Mahieu M, Elmer B, Bretteville A, Wang Y, Lopez-Grancha M, Goniot P, Moindrot N, Ferrari P, Blanc V, Schussler N, Brault E, Roudières V, Blanchard V, Yang ZY, Barneoud P, Bertrand P, Roucourt B, Carmans S, Bottelbergs A, Mertens L, Wintmolders C, Larsen P, Hersley C, McGathey T, Racke MM, Liu L, Lu J, O'Neill MJ, Riddell DR, Ebneth A, Nabel GJ, Pradier L. Systemic immune-checkpoint blockade with anti-PD1 antibodies does not alter cerebral amyloid-β burden in several amyloid transgenic mouse models. Glia 2017; 66:492-504. [PMID: 29134678 DOI: 10.1002/glia.23260] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 10/20/2017] [Accepted: 10/24/2017] [Indexed: 01/22/2023]
Abstract
Chronic inflammation represents a central component in the pathogenesis of Alzheimer's disease (AD). Recent work suggests that breaking immune tolerance by Programmed cell Death-1 (PD1) checkpoint inhibition produces an IFN-γ-dependent systemic immune response, with infiltration of the brain by peripheral myeloid cells and neuropathological as well as functional improvements even in mice with advanced amyloid pathology (Baruch et al., (): Nature Medicine, 22:135-137). Immune checkpoint inhibition was therefore suggested as potential treatment for neurodegenerative disorders when activation of the immune system is appropriate. Because a xenogeneic rat antibody (mAb) was used in the study, whether the effect was specific to PD1 target engagement was uncertain. In the present study we examined whether PD1 immunotherapy can lower amyloid-β pathology in a range of different amyloid transgenic models performed at three pharmaceutical companies with the exact same anti-PD1 isotype and two mouse chimeric variants. Although PD1 immunotherapy stimulated systemic activation of the peripheral immune system, monocyte-derived macrophage infiltration into the brain was not detected, and progression of brain amyloid pathology was not altered. Similar negative results of the effect of PD1 immunotherapy on amyloid brain pathology were obtained in two additional models in two separate institutions. These results show that inhibition of PD1 checkpoint signaling by itself is not sufficient to reduce amyloid pathology and that additional factors might have contributed to previously published results (Baruch et al., (): Nature Medicine, 22:135-137). Until such factors are elucidated, animal model data do not support further evaluation of PD1 checkpoint inhibition as a therapeutic modality for Alzheimer's disease.
Collapse
Affiliation(s)
| | - Bradford Elmer
- SANOFI NA BT Lab, 270 Albany St, Cambridge, Massachusetts, 02319
| | - Alexis Bretteville
- Janssen Research & Development, a Division of Janssen Pharmaceutica N.V, Beerse, B-2340, Belgium
| | - Yaming Wang
- Lilly Research Laboratories, Indianapolis, Indiana, 46285
| | - Mati Lopez-Grancha
- SANOFI Neurosciences, 1 rue P. Brossolette, Chilly-Mazarin, 91385, France
| | - Philippe Goniot
- SANOFI Neurosciences, 1 rue P. Brossolette, Chilly-Mazarin, 91385, France
| | - Nicolas Moindrot
- SANOFI Neurosciences, 1 rue P. Brossolette, Chilly-Mazarin, 91385, France
| | - Paul Ferrari
- SANOFI, 13 quai Jules Guesde, Vitry/Seine, 94403, France
| | | | - Nathalie Schussler
- SANOFI Neurosciences, 1 rue P. Brossolette, Chilly-Mazarin, 91385, France
| | - Emmanuel Brault
- SANOFI Neurosciences, 1 rue P. Brossolette, Chilly-Mazarin, 91385, France
| | - Valérie Roudières
- SANOFI Neurosciences, 1 rue P. Brossolette, Chilly-Mazarin, 91385, France
| | | | - Zhi-Yong Yang
- SANOFI NA BT Lab, 270 Albany St, Cambridge, Massachusetts, 02319
| | - Pascal Barneoud
- SANOFI Neurosciences, 1 rue P. Brossolette, Chilly-Mazarin, 91385, France
| | - Philippe Bertrand
- SANOFI Neurosciences, 1 rue P. Brossolette, Chilly-Mazarin, 91385, France
| | - Bart Roucourt
- reMYND NV, Gaston Geenslaan 1, Leuven, 3001, Belgium
| | - Sofie Carmans
- reMYND NV, Gaston Geenslaan 1, Leuven, 3001, Belgium
| | - Astrid Bottelbergs
- Janssen Research & Development, a Division of Janssen Pharmaceutica N.V, Beerse, B-2340, Belgium
| | - Liesbeth Mertens
- Janssen Research & Development, a Division of Janssen Pharmaceutica N.V, Beerse, B-2340, Belgium
| | - Cindy Wintmolders
- Janssen Research & Development, a Division of Janssen Pharmaceutica N.V, Beerse, B-2340, Belgium
| | - Peter Larsen
- Janssen Research & Development, a Division of Janssen Pharmaceutica N.V, Beerse, B-2340, Belgium
| | | | - Tyler McGathey
- Lilly Research Laboratories, Indianapolis, Indiana, 46285
| | | | - Ling Liu
- Lilly Research Laboratories, Indianapolis, Indiana, 46285
| | - Jirong Lu
- Lilly Research Laboratories, Indianapolis, Indiana, 46285
| | | | | | - Andreas Ebneth
- Janssen Research & Development, a Division of Janssen Pharmaceutica N.V, Beerse, B-2340, Belgium
| | - Gary J Nabel
- SANOFI NA BT Lab, 270 Albany St, Cambridge, Massachusetts, 02319
| | - Laurent Pradier
- SANOFI Neurosciences, 1 rue P. Brossolette, Chilly-Mazarin, 91385, France
| |
Collapse
|
24
|
Mast N, Saadane A, Valencia-Olvera A, Constans J, Maxfield E, Arakawa H, Li Y, Landreth G, Pikuleva IA. Cholesterol-metabolizing enzyme cytochrome P450 46A1 as a pharmacologic target for Alzheimer's disease. Neuropharmacology 2017; 123:465-476. [PMID: 28655608 PMCID: PMC5546235 DOI: 10.1016/j.neuropharm.2017.06.026] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 06/21/2017] [Accepted: 06/23/2017] [Indexed: 12/27/2022]
Abstract
Cytochrome P450 46A1 (CYP46A1 or cholesterol 24-hydroxylase) controls cholesterol elimination from the brain and plays a role in higher order brain functions. Genetically enhanced CYP46A1 expression in mouse models of Alzheimer's disease mitigates the manifestations of this disease. We enhanced CYP46A1 activity pharmacologically by treating 5XFAD mice, a model of rapid amyloidogenesis, with a low dose of the anti-HIV medication efavirenz. Efavirenz was administered from 1 to 9 months of age, and mice were evaluated at specific time points. At one month of age, cholesterol homeostasis was already disturbed in the brain of 5XFAD mice. Nevertheless, efavirenz activated CYP46A1 and mouse cerebral cholesterol turnover during the first four months of administration. This treatment time also reduced amyloid burden and microglia activation in the cortex and subiculum of 5XFAD mice as well as protein levels of amyloid precursor protein and the expression of several genes involved in inflammatory response. However, mouse short-term memory and long-term spatial memory were impaired, whereas learning in the context-dependent fear test was improved. Additional four months of drug administration (a total of eight months of treatment) improved long-term spatial memory in the treated as compared to the untreated mice, further decreased amyloid-β content in 5XFAD brain, and also decreased the mortality rate among male mice. We propose a mechanistic model unifying the observed efavirenz effects. We suggest that CYP46A1 activation by efavirenz could be a new anti-Alzheimer's disease treatment and a tool to study and identify normal and pathological brain processes affected by cholesterol maintenance.
Collapse
Affiliation(s)
- Natalia Mast
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Aicha Saadane
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Ana Valencia-Olvera
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - James Constans
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Erin Maxfield
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Hiroyuki Arakawa
- Behavioral Core, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Young Li
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Gary Landreth
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Irina A Pikuleva
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH 44106, USA.
| |
Collapse
|
25
|
Hui L, Han M, Du XD, Zhang BH, He SC, Shao TN, Yin GZ. Serum ApoB levels in depressive patients: associated with cognitive deficits. Sci Rep 2017; 7:39992. [PMID: 28054633 PMCID: PMC5215543 DOI: 10.1038/srep39992] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 11/30/2016] [Indexed: 11/17/2022] Open
Abstract
Cognitive deficits have been regarded as one of the most significant clinical symptoms of depressive disorder. Accumulating evidence has shown that apolipoprotein B (ApoB) levels, which are responsible for inducing neurodegeneration, may be involved in cognitive deficits. This study examines cognitive deficits, and the correlation of serum ApoB levels with cognitive deficits of depressive disorder. 90 depressive patients and 90 healthy controls with matched age and gender were recruited. Cognition was assessed using the Repeatable Battery for the Assessment of Neuropsychological Status (RBANS). Serum ApoB levels in depressive patients were measured by immunoturbidimetric method. Our results showed that depressive patients had lower scores of cognition including RBANS total score and subscales of language and delayed memory (all, p < 0.001) than healthy controls after controlling for the variables. The differences in cognitive functions also passed Bonferroni corrections. Serum ApoB levels were negatively correlated with delayed memory score in depressive patients (r = −0.30, p = 0.01). Furthermore, stepwise multivariate regression analysis indicated that serum ApoB levels independently contributed to delayed memory in depressive patients (t = −2.68, p = 0.01). Our findings support that serum ApoB levels may be involved in delayed memory decline in depressive patients. Depressive patients also experience greater cognitive deficits, especially in delayed memory and language than healthy controls.
Collapse
Affiliation(s)
- Li Hui
- Institute of Mental Health, Suzhou Psychiatric Hospital, The Affiliated Guangji Hospital of Soochow University, Suzhou 215008, Jiangsu, PR China
| | - Mei Han
- School of Medicine, IHMRI, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Xiang Dong Du
- Institute of Mental Health, Suzhou Psychiatric Hospital, The Affiliated Guangji Hospital of Soochow University, Suzhou 215008, Jiangsu, PR China
| | - Bao Hua Zhang
- Beijing HuilongGuan Hospital, Peking University, Beijing 100096, PR China
| | - Shu Chang He
- Department of Psychology, Peking University, Beijing 100000, PR China
| | - Tian Nan Shao
- Institute of Mental Health, Suzhou Psychiatric Hospital, The Affiliated Guangji Hospital of Soochow University, Suzhou 215008, Jiangsu, PR China
| | - Guang Zhong Yin
- Institute of Mental Health, Suzhou Psychiatric Hospital, The Affiliated Guangji Hospital of Soochow University, Suzhou 215008, Jiangsu, PR China
| |
Collapse
|
26
|
Löffler T, Schweinzer C, Flunkert S, Sántha M, Windisch M, Steyrer E, Hutter-Paier B. Brain cortical cholesterol metabolism is highly affected by human APP overexpression in mice. Mol Cell Neurosci 2016; 74:34-41. [DOI: 10.1016/j.mcn.2016.03.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 03/07/2016] [Accepted: 03/14/2016] [Indexed: 12/27/2022] Open
|
27
|
Lénárt N, Walter FR, Bocsik A, Sántha P, Tóth ME, Harazin A, Tóth AE, Vizler C, Török Z, Pilbat AM, Vígh L, Puskás LG, Sántha M, Deli MA. Cultured cells of the blood-brain barrier from apolipoprotein B-100 transgenic mice: effects of oxidized low-density lipoprotein treatment. Fluids Barriers CNS 2015; 12:17. [PMID: 26184769 PMCID: PMC4504453 DOI: 10.1186/s12987-015-0013-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 07/01/2015] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND The apolipoprotein B-100 (ApoB-100) transgenic mouse line is a model of human atherosclerosis. Latest findings suggest the importance of ApoB-100 in the development of neurodegenerative diseases and microvascular/perivascular localization of ApoB-100 protein was demonstrated in the cerebral cortex of ApoB-100 transgenic mice. The aim of the study was to characterize cultured brain endothelial cells, pericytes and glial cells from wild-type and ApoB-100 transgenic mice and to study the effect of oxidized low-density lipoprotein (oxLDL) on these cells. METHODS Morphology of cells isolated from brains of wild type and ApoB-100 transgenic mice was characterized by immunohistochemistry and the intensity of immunolabeling was quantified by image analysis. Toxicity of oxLDL treatment was monitored by real-time impedance measurement and lactate dehydrogenase release. Reactive oxygen species and nitric oxide production, barrier permeability in triple co-culture blood-brain barrier model and membrane fluidity were also determined after low-density lipoprotein (LDL) or oxLDL treatment. RESULTS The presence of ApoB-100 was confirmed in brain endothelial cells, while no morphological change was observed between wild type and transgenic cells. Oxidized but not native LDL exerted dose-dependent toxicity in all three cell types, induced barrier dysfunction and increased reactive oxygen species (ROS) production in both genotypes. A partial protection from oxLDL toxicity was seen in brain endothelial and glial cells from ApoB-100 transgenic mice. Increased membrane rigidity was measured in brain endothelial cells from ApoB-100 transgenic mice and in LDL or oxLDL treated wild type cells. CONCLUSION The morphological and functional properties of cultured brain endothelial cells, pericytes and glial cells from ApoB-100 transgenic mice were characterized and compared to wild type cells for the first time. The membrane fluidity changes in ApoB-100 transgenic cells related to brain microvasculature indicate alterations in lipid composition which may be linked to the partial protection against oxLDL toxicity.
Collapse
Affiliation(s)
- Nikolett Lénárt
- Laboratory of Animal Genetics and Molecular Neurobiology, Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Temesvári krt. 62, 6720, Szeged, Hungary. .,Laboratory of Neuroimmunology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary.
| | - Fruzsina R Walter
- Biological Barriers Research Group, Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary.
| | - Alexandra Bocsik
- Biological Barriers Research Group, Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary.
| | - Petra Sántha
- Biological Barriers Research Group, Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary.
| | - Melinda E Tóth
- Laboratory of Animal Genetics and Molecular Neurobiology, Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Temesvári krt. 62, 6720, Szeged, Hungary.
| | - András Harazin
- Biological Barriers Research Group, Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary.
| | - Andrea E Tóth
- Biological Barriers Research Group, Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary.
| | - Csaba Vizler
- Laboratory of Molecular Stress Biology, Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary.
| | - Zsolt Török
- Laboratory of Molecular Stress Biology, Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary.
| | - Ana-Maria Pilbat
- Laboratory of Molecular Stress Biology, Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary.
| | - László Vígh
- Laboratory of Molecular Stress Biology, Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary.
| | - László G Puskás
- Laboratory of Functional Genomics, Laboratories of Core Facilities, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary.
| | - Miklós Sántha
- Laboratory of Animal Genetics and Molecular Neurobiology, Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Temesvári krt. 62, 6720, Szeged, Hungary.
| | - Mária A Deli
- Biological Barriers Research Group, Institute of Biophysics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary.
| |
Collapse
|
28
|
Krassnig S, Schweinzer C, Taub N, Havas D, Auer E, Flunkert S, Schreibmayer W, Hutter-Paier B, Windisch M. Influence of Lentiviral β-Synuclein Overexpression in the Hippocampus of a Transgenic Mouse Model of Alzheimer's Disease on Amyloid Precursor Protein Metabolism and Pathology. NEURODEGENER DIS 2015; 15:243-57. [PMID: 26111745 DOI: 10.1159/000430952] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Accepted: 04/26/2015] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND β-Synuclein (β-Syn) is a member of the highly homologous synuclein protein family. The most prominent family member, α-synuclein (α-Syn), abnormally accumulates in so-called Lewy bodies, one of the major pathological hallmarks of α-synucleinopathies. Notably, parts of the peptide backbone, called the nonamyloid component, are also found in amyloid plaques. However, β-Syn seems to have beneficial effects by reducing α-Syn aggregation, and amyloid antiaggregatory activity has been described. OBJECTIVE The aim of the study was to analyze if wild-type β-Syn can counteract functional and pathological changes in a murine Alzheimer model over different time periods. METHODS At the onset of pathology, lentiviral particles expressing human β-Syn were injected into the hippocampus of transgenic mice overexpressing human amyloid precursor protein with Swedish and London mutations (APPSL). An empty vector served as the control. Behavioral analyses were performed 1, 3 and 6 months after injection followed by biochemical and histological examinations of brain samples. RESULTS β-Syn expression was locally concentrated and rather modest, but nevertheless changed its effect on APP expression and plaque load in a time- and concentration-dependent manner. Interestingly, the phosphorylation of glycogen synthase kinase 3 beta was enhanced in APPSL mice expressing human β-Syn, but an inverse trend was observed in wild-type animals. CONCLUSION The initially reported beneficial effects of β-Syn could be partially reproduced, but locally elevated levels of β-Syn might also cause neurodegeneration. To enlighten the controversial pathological mechanism of β-Syn, further examinations considering the relationship between concentration and exposure time of β-Syn are needed.
Collapse
|
29
|
Yamchuen P, Aimjongjun S, Limpeanchob N. Oxidized low density lipoprotein increases acetylcholinesterase activity correlating with reactive oxygen species production. Neurochem Int 2014; 78:1-6. [DOI: 10.1016/j.neuint.2014.07.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 06/30/2014] [Accepted: 07/24/2014] [Indexed: 01/08/2023]
|
30
|
Löffler T, Flunkert S, Havas D, Schweinzer C, Uger M, Windisch M, Steyrer E, Hutter-Paier B. Neuroinflammation and related neuropathologies in APPSL mice: further value of this in vivo model of Alzheimer's disease. J Neuroinflammation 2014; 11:84. [PMID: 24886182 PMCID: PMC4108132 DOI: 10.1186/1742-2094-11-84] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Accepted: 04/10/2014] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Beyond cognitive decline, Alzheimer's disease (AD) is characterized by numerous neuropathological changes in the brain. Although animal models generally do not fully reflect the broad spectrum of disease-specific alterations, the APPSL mouse model is well known to display early plaque formation and to exhibit spatial learning and memory deficits. However, important neuropathological features, such as neuroinflammation and lipid peroxidation, and their progression over age, have not yet been described in this AD mouse model. METHODS Hippocampal and neocortical tissues of APPSL mice at different ages were evaluated. One hemisphere from each mouse was examined for micro- and astrogliosis as well as concomitant plaque load. The other hemisphere was evaluated for lipid peroxidation (quantified by a thiobarbituric acid reactive substances (TBARS) assay), changes in Aβ abundance (Aβ38, Aβ40 and Aβ42 analyses), as well as determination of aggregated Aβ content (Amorfix A4 assay). Finally, correlation analyses were performed to illustrate the time-dependent correlation between neuroinflammation and Aβ load (soluble, insoluble, fibrils), or lipid peroxidation, respectively. RESULTS As is consistent with previous findings, neuroinflammation starts early and shows strong progression over age in the APPSL mouse model. An analyses of concomitant Aβ load and plaque deposition revealed a similar progression, and high correlations between neuroinflammation markers and soluble or insoluble Aβ or fibrillar amyloid plaque loads were observed. Lipid peroxidation, as measured by TBARS levels, correlates well with neuroinflammation in the neocortex but not the hippocampus. The hippocampal lipid peroxidation correlated strongly with the increase of LOC positive fiber load, whereas neocortical TBARS levels were unrelated to amyloidosis. CONCLUSIONS These data illustrate for the first time the progression of major AD related neuropathological features other than plaque load in the APPSL mouse model. Specifically, we demonstrate that microgliosis and astrocytosis are prominent aspects of this AD mouse model. The strong correlation of neuroinflammation with amyloid burden and lipid peroxidation underlines the importance of these pathological factors for the development of AD. The new finding of a different relation of lipid peroxidation in the hippocampus and neocortical regions show that the model might contribute to the understanding of complex pathological mechanisms and their interplay in AD.
Collapse
Affiliation(s)
- Tina Löffler
- QPS-Austria GmbH, Parkring 12, 8074 Grambach, Austria
- Institute of Molecular Biology and Biochemistry, Medical University Graz, Harrachgasse 21, 8010 Graz, Austria
| | | | - Daniel Havas
- QPS-Austria GmbH, Parkring 12, 8074 Grambach, Austria
| | | | - Marni Uger
- Amorfix Life Sciences Ltd, 3403 American Drive, Ontario, Canada L4V 1 T4
| | | | - Ernst Steyrer
- Institute of Molecular Biology and Biochemistry, Medical University Graz, Harrachgasse 21, 8010 Graz, Austria
| | | |
Collapse
|
31
|
Windisch M. We Can Treat Alzheimer's Disease Successfully in Mice but Not in Men: Failure in Translation? A Perspective. NEURODEGENER DIS 2014; 13:147-50. [DOI: 10.1159/000357568] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 11/22/2013] [Indexed: 11/19/2022] Open
|
32
|
Chen X, Hui L, Geiger JD. Role of LDL cholesterol and endolysosomes in amyloidogenesis and Alzheimer's disease. ACTA ACUST UNITED AC 2014; 5. [PMID: 26413387 DOI: 10.4172/2155-9562.1000236] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The pathogenesis of late-onset sporadic Alzheimer's disease (AD) is believed to result from complex interactions between nutritional, environmental, epigenetic and genetic factors. Among those factors, altered circulating cholesterol homeostasis, independent of the APOE genotype, continues to be implicated in brain deposition of amyloid beta protein (Aβ) and the pathogenesis of AD. It is believed that trafficking of amyloid beta precursor protein (AβPP) into endolysosomes appears to play a critical role in determining amyloidogenic processing of AβPP because this is precisely where two enzymes critically important in AβPP metabolism are located; beta amyloid converting enzyme (BACE-1) and gamma secretase enzyme. We have shown that elevated levels of LDL cholesterol promote AβPP internalization, disturb neuronal endolysosome structure and function, and increase Aβ accumulation in neuronal endolysosomes. Here, we will further discuss the linkage between elevated levels of LDL cholesterol and AD pathogenesis, and explore the underlying mechanisms whereby elevated levels of plasma LDL cholesterol promote amyloidogenesis.
Collapse
Affiliation(s)
- Xuesong Chen
- Department of Basic Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203
| | - Liang Hui
- Department of Basic Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203
| | - Jonathan D Geiger
- Department of Basic Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203
| |
Collapse
|