1
|
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 2024:100667. [PMID: 39395793 DOI: 10.1016/j.jlr.2024.100667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 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 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, Montréal, Québec, Canada, H4H 1R3; Centre for the Studies in the Prevention of Alzheimer's Disease, Montréal, Québec, Canada, H4H 1R3; McGill University, Montréal, Québec, Canada, H3A 0G4
| | - Cynthia Picard
- Douglas Mental Health University Institute, Montréal, Québec, Canada, H4H 1R3; Centre for the Studies in the Prevention of Alzheimer's Disease, Montréal, Québec, Canada, H4H 1R3
| | - Anne Labonté
- Douglas Mental Health University Institute, Montréal, Québec, Canada, H4H 1R3; Centre for the Studies in the Prevention of Alzheimer's Disease, Montréal, Québec, Canada, H4H 1R3
| | - Judes Poirier
- Douglas Mental Health University Institute, Montréal, Québec, Canada, H4H 1R3; Centre for the Studies in the Prevention of Alzheimer's Disease, Montréal, Québec, Canada, H4H 1R3; McGill University, Montréal, Québec, Canada, H3A 0G4
| |
Collapse
|
2
|
Pavešković M, De-Paula RB, Ojelade SA, Tantry EK, Kochukov MY, Bao S, Veeraragavan S, Garza AR, Srivastava S, Song SY, Fujita M, Duong DM, Bennett DA, De Jager PL, Seyfried NT, Dickinson ME, Heaney JD, Arenkiel BR, Shulman JM. Alzheimer's disease risk gene CD2AP is a dose-sensitive determinant of synaptic structure and plasticity. Hum Mol Genet 2024; 33:1815-1832. [PMID: 39146503 PMCID: PMC11458016 DOI: 10.1093/hmg/ddae115] [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: 02/27/2024] [Revised: 06/15/2024] [Indexed: 08/17/2024] Open
Abstract
CD2-Associated protein (CD2AP) is a candidate susceptibility gene for Alzheimer's disease, but its role in the mammalian central nervous system remains largely unknown. We show that CD2AP protein is broadly expressed in the adult mouse brain, including within cortical and hippocampal neurons, where it is detected at pre-synaptic terminals. Deletion of Cd2ap altered dendritic branching and spine density, and impaired ubiquitin-proteasome system activity. Moreover, in mice harboring either one or two copies of a germline Cd2ap null allele, we noted increased paired-pulse facilitation at hippocampal Schaffer-collateral synapses, consistent with a haploinsufficient requirement for pre-synaptic release. Whereas conditional Cd2ap knockout in the brain revealed no gross behavioral deficits in either 3.5- or 12-month-old mice, Cd2ap heterozygous mice demonstrated subtle impairments in discrimination learning using a touchscreen task. Based on unbiased proteomics, partial or complete loss of Cd2ap triggered perturbation of proteins with roles in protein folding, lipid metabolism, proteostasis, and synaptic function. Overall, our results reveal conserved, dose-sensitive requirements for CD2AP in the maintenance of neuronal structure and function, including synaptic homeostasis and plasticity, and inform our understanding of possible cell-type specific mechanisms in Alzheimer's Disease.
Collapse
Affiliation(s)
- Matea Pavešković
- Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, 1250 Moursund Street, Houston, TX 77030, United States
- Department of Neurology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
| | - Ruth B De-Paula
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, 1250 Moursund Street, Houston, TX 77030, United States
- Department of Neurology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
- Quantitative and Computational Biology Program, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
| | - Shamsideen A Ojelade
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, 1250 Moursund Street, Houston, TX 77030, United States
- Department of Neurology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
| | - Evelyne K Tantry
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, 1250 Moursund Street, Houston, TX 77030, United States
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
| | - Mikhail Y Kochukov
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, 1250 Moursund Street, Houston, TX 77030, United States
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
| | - Suyang Bao
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, 1250 Moursund Street, Houston, TX 77030, United States
- Development, Disease Models, and Therapeutics Graduate Program, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
| | - Surabi Veeraragavan
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, 1250 Moursund Street, Houston, TX 77030, United States
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
| | - Alexandra R Garza
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, 1250 Moursund Street, Houston, TX 77030, United States
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
| | - Snigdha Srivastava
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, 1250 Moursund Street, Houston, TX 77030, United States
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
- Medical Scientist Training Program, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
| | - Si-Yuan Song
- Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, 1250 Moursund Street, Houston, TX 77030, United States
| | - Masashi Fujita
- Center for Translational and Computational Neuroimmunology, Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, 630 West 168th Street, New York, NY, United States
| | - Duc M Duong
- Departments of Biochemistry and Neurology, Emory University School of Medicine, 100 Woodruff Circle, Atlanta, GA 30322, United States
| | - David A Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, 600 S. Paulina Street, Chicago, IL 60612, United States
| | - Philip L De Jager
- Center for Translational and Computational Neuroimmunology, Department of Neurology and the Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, 630 West 168th Street, New York, NY, United States
| | - Nicholas T Seyfried
- Departments of Biochemistry and Neurology, Emory University School of Medicine, 100 Woodruff Circle, Atlanta, GA 30322, United States
| | - Mary E Dickinson
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
| | - Jason D Heaney
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
| | - Benjamin R Arenkiel
- Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, 1250 Moursund Street, Houston, TX 77030, United States
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
| | - Joshua M Shulman
- Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, 1250 Moursund Street, Houston, TX 77030, United States
- Department of Neurology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
- Center for Alzheimer’s and Neurodegenerative Diseases, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, United States
| |
Collapse
|
3
|
Zhang D, Wu J, Ren G, Wang Y, Xu H, Chen S, Li X, Chen X. Diagnostic Value of Serum Apolipoprotein B100 Combined With Hippocampal Volume in Alzheimer's Disease. Brain Behav 2024; 14:e70066. [PMID: 39344486 PMCID: PMC11440087 DOI: 10.1002/brb3.70066] [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: 12/28/2023] [Revised: 08/22/2024] [Accepted: 09/08/2024] [Indexed: 10/01/2024] Open
Abstract
PURPOSE To explore the diagnostic value of serum apolipoprotein B100 (Apo B100) combined with hippocampal volume in Alzheimer's disease (AD). METHODS A total of 59 AD patients and 59 healthy subjects were selected. The Mini-Mental State Examination (MMSE) was used for neuropsychological assessment. Blood glucose and serum lipid levels were detected by biochemical analyzer. Polymerase chain reaction (PCR) was used to detect apolipoprotein E (Apo E) ε3/ε4 genotypes in the plasma. Hippocampal volume was calculated using Slicer software. Independent-sample t test or Mann-Whitney U test were used to compare the levels of various indicators between the two groups. Spearman's correlation analysis was used to analyze the correlation between each level. The receiver operating characteristic curve (ROC) was plotted, and the area under the curve (AUC) was calculated to compare the diagnostic efficacy of individual and combined detection of serum Apo B100 levels and hippocampal volume in AD. RESULTS Compared with the healthy control group, the levels of serum total cholesterol (TC), low-density lipoprotein (LDL), Apo B100, and plasma Apo E ε3/ε4 were higher in the AD group, and serum high-density lipoprotein (HDL) level was lower in the AD group (both p < 0.05). The hippocampal volume in the AD group was lower than in the control group (p < 0.01). The serum Apo B100 level was negatively correlated with MMSE score (r = -0.646), whereas hippocampal volume was positively correlated with MMSE score (r = 0.630). ROC curve analysis showed that the AUC of the combined serum Apo B100 level and hippocampal volume for AD was higher than that of either alone (AUC = 0.821, p < 0.01). CONCLUSION Serum Apo B100 level is elevated, and the hippocampal volume is reduced in AD patients. The combined detection of the two has a higher diagnostic efficiency for AD than other alone and has the potential to become an important indicator for the diagnosis of AD in the future.
Collapse
Affiliation(s)
- Dandan Zhang
- Department of NeurologyThe Affiliated People's Hospital ofJiangsu UniversityZhenjiangJiangsuPeople's Republic of China
| | - Jing Wu
- Department of NeurologyThe Affiliated People's Hospital ofJiangsu UniversityZhenjiangJiangsuPeople's Republic of China
| | - Guoqiang Ren
- Department of RadiologyThe Affiliated People's Hospital of Jiangsu UniversityZhenjiangJiangsuPeople's Republic of China
| | - Yi Wang
- Department of NeurologyThe People's Hospital of Jurong CityJurongJiangsuPeople's Republic of China
| | - Hang Xu
- Department of NeurologyThe Affiliated People's Hospital ofJiangsu UniversityZhenjiangJiangsuPeople's Republic of China
| | - Siyuan Chen
- Department of NeurologyThe Affiliated People's Hospital ofJiangsu UniversityZhenjiangJiangsuPeople's Republic of China
| | - Xuezhong Li
- Department of NeurologyThe Affiliated People's Hospital ofJiangsu UniversityZhenjiangJiangsuPeople's Republic of China
| | - Xiaopeng Chen
- Department of NeurologyThe Affiliated People's Hospital ofJiangsu UniversityZhenjiangJiangsuPeople's Republic of China
| |
Collapse
|
4
|
Ding M, Li QF, Peng TH, Wang TQ, Yan HH, Tang C, Wang XY, Guo Y, Zheng L. Early life exercise training and inhibition of apoLpp mRNA expression to improve age-related arrhythmias and prolong the average lifespan in Drosophila melanogaster. Aging (Albany NY) 2022; 14:9908-9923. [PMID: 36470666 PMCID: PMC9831727 DOI: 10.18632/aging.204422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 11/16/2022] [Indexed: 01/03/2023]
Abstract
Cardiovascular disease (CVD) places a heavy burden on older patients and the global healthcare system. A large body of evidence suggests that exercise training is essential in preventing and treating cardiovascular disease, but the underlying mechanisms are not well understood. Here, we used the Drosophila melanogaster animal model to study the effects of early-life exercise training (Exercise) on the aging heart and lifespan. We found in flies that age-induced arrhythmias are conserved across different genetic backgrounds. The fat body is the primary source of circulating lipoproteins in flies. Inhibition of fat body apoLpp (Drosophila apoB homolog) demonstrated that low expression of apoLpp reduced the development of arrhythmias in aged flies but did not affect average lifespan. At the same time, exercise can also reduce the expression of apoLpp mRNA in aged flies and have a protective effect on the heart, which is similar to the inhibition of apoLpp mRNA. Although treatment of UAS-apoLppRNAi and exercise alone had no significant effect on lifespan, the combination of UAS-apoLppRNAi and exercise extended the average lifespan of flies. Therefore, we conclude that UAS-apoLppRNAi and exercise are sufficient to resist age-induced arrhythmias, which may be related to the decreased expression of apoLpp mRNA, and that UAS-apoLppRNAi and exercise have a combined effect on prolonging the average lifespan.
Collapse
Affiliation(s)
- Meng Ding
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, Hunan Normal University, Changsha, China
| | - Qiu Fang Li
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, Hunan Normal University, Changsha, China
| | - Tian Hang Peng
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, Hunan Normal University, Changsha, China
| | - Tong Quan Wang
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, Hunan Normal University, Changsha, China
| | - Han Hui Yan
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, Hunan Normal University, Changsha, China
| | - Chao Tang
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, Hunan Normal University, Changsha, China
| | - Xiao Ya Wang
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, Hunan Normal University, Changsha, China
| | - Yin Guo
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, Hunan Normal University, Changsha, China
| | - Lan Zheng
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, Hunan Normal University, Changsha, China
| |
Collapse
|
5
|
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: 0] [Impact Index Per Article: 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
|
6
|
Tóth ME, Sárközy M, Szűcs G, Dukay B, Hajdu P, Zvara Á, Puskás LG, Szebeni GJ, Ruppert Z, Csonka C, Kovács F, Kriston A, Horváth P, Kővári B, Cserni G, Csont T, Sántha M. Exercise training worsens cardiac performance in males but does not change ejection fraction and improves hypertrophy in females in a mouse model of metabolic syndrome. Biol Sex Differ 2022; 13:5. [PMID: 35101146 PMCID: PMC8805345 DOI: 10.1186/s13293-022-00414-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 01/05/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Metabolic syndrome (MetS) refers to a cluster of co-existing cardio-metabolic risk factors, including visceral obesity, dyslipidemia, hyperglycemia with insulin resistance, and hypertension. As there is a close link between MetS and cardiovascular diseases, we aimed to investigate the sex-based differences in MetS-associated heart failure (HF) and cardiovascular response to regular exercise training (ET). METHODS High-fat diet-fed male and female APOB-100 transgenic (HFD/APOB-100, 3 months) mice were used as MetS models, and age- and sex-matched C57BL/6 wild-type mice on standard diet served as healthy controls (SD/WT). Both the SD/WT and HFD/APOB-100 mice were divided into sedentary and ET groups, the latter running on a treadmill (0.9 km/h) for 45 min 5 times per week for 7 months. At month 9, transthoracic echocardiography was performed to monitor cardiac function and morphology. At the termination of the experiment at month 10, blood was collected for serum low-density lipoprotein (LDL)- and high-density lipoprotein (HDL)-cholesterol measurements and homeostatic assessment model for insulin resistance (HOMA-IR) calculation. Cardiomyocyte hypertrophy and fibrosis were assessed by histology. Left ventricular expressions of selected genes associated with metabolism, inflammation, and stress response were investigated by qPCR. RESULTS Both HFD/APOB-100 males and females developed obesity and hypercholesterolemia; however, only males showed insulin resistance. ET did not change these metabolic parameters. HFD/APOB-100 males showed echocardiographic signs of mild HF with dilated ventricles and thinner walls, whereas females presented the beginning of left ventricular hypertrophy. In response to ET, SD/WT males developed increased left ventricular volumes, whereas females responded with physiologic hypertrophy. Exercise-trained HFD/APOB-100 males presented worsening HF with reduced ejection fraction; however, ET did not change the ejection fraction and reversed the echocardiographic signs of left ventricular hypertrophy in HFD/APOB-100 females. The left ventricular expression of the leptin receptor was higher in females than males in the SD/WT groups. Left ventricular expression levels of stress response-related genes were higher in the exercise-trained HFD/APOB-100 males and exercise-trained SD/WT females than exercise-trained SD/WT males. CONCLUSIONS HFD/APOB-100 mice showed sex-specific cardiovascular responses to MetS and ET; however, left ventricular gene expressions were similar between the groups except for leptin receptor and several stress response-related genes.
Collapse
Affiliation(s)
- Melinda E. Tóth
- grid.481814.00000 0004 0479 9817Laboratory of Animal Genetics and Molecular Neurobiology, Institute of Biochemistry, Biological Research Centre, Eötvös Loránd Research Network, Temesvári krt. 62, Szeged, 6726 Hungary
| | - Márta Sárközy
- MEDICS Research Group, Department of Biochemistry, University of Szeged Albert Szent-Györgyi Medical School, Dóm tér 9, Szeged, 6720, Hungary. .,Interdisciplinary Center of Excellence, University of Szeged, Dugonics tér 13, Szeged, 6720, Hungary.
| | - Gergő Szűcs
- grid.9008.10000 0001 1016 9625MEDICS Research Group, Department of Biochemistry, University of Szeged Albert Szent-Györgyi Medical School, Dóm tér 9, Szeged, 6720 Hungary ,grid.9008.10000 0001 1016 9625Interdisciplinary Center of Excellence, University of Szeged, Dugonics tér 13, Szeged, 6720 Hungary
| | - Brigitta Dukay
- grid.481814.00000 0004 0479 9817Laboratory of Animal Genetics and Molecular Neurobiology, Institute of Biochemistry, Biological Research Centre, Eötvös Loránd Research Network, Temesvári krt. 62, Szeged, 6726 Hungary
| | - Petra Hajdu
- grid.481814.00000 0004 0479 9817Laboratory of Animal Genetics and Molecular Neurobiology, Institute of Biochemistry, Biological Research Centre, Eötvös Loránd Research Network, Temesvári krt. 62, Szeged, 6726 Hungary
| | - Ágnes Zvara
- grid.418331.c0000 0001 2195 9606Laboratory of Functional Genomics, Biological Research Centre, Eötvös Loránd Research Network, Temesvári krt. 62, Szeged, 6726 Hungary
| | - László G. Puskás
- grid.418331.c0000 0001 2195 9606Laboratory of Functional Genomics, Biological Research Centre, Eötvös Loránd Research Network, Temesvári krt. 62, Szeged, 6726 Hungary
| | - Gábor J. Szebeni
- grid.418331.c0000 0001 2195 9606Laboratory of Functional Genomics, Biological Research Centre, Eötvös Loránd Research Network, Temesvári krt. 62, Szeged, 6726 Hungary
| | - Zsófia Ruppert
- grid.481814.00000 0004 0479 9817Laboratory of Animal Genetics and Molecular Neurobiology, Institute of Biochemistry, Biological Research Centre, Eötvös Loránd Research Network, Temesvári krt. 62, Szeged, 6726 Hungary ,grid.9008.10000 0001 1016 9625Doctoral School in Biology, University of Szeged, Szeged, Hungary
| | - Csaba Csonka
- grid.9008.10000 0001 1016 9625MEDICS Research Group, Department of Biochemistry, University of Szeged Albert Szent-Györgyi Medical School, Dóm tér 9, Szeged, 6720 Hungary ,grid.9008.10000 0001 1016 9625Interdisciplinary Center of Excellence, University of Szeged, Dugonics tér 13, Szeged, 6720 Hungary
| | - Ferenc Kovács
- grid.481814.00000 0004 0479 9817Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Eötvös Loránd Research Network, Temesvári krt. 62, Szeged, 6726 Hungary ,Single-Cell Technologies Ltd, Temesvári krt. 62, Szeged, 6726 Hungary
| | - András Kriston
- grid.481814.00000 0004 0479 9817Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Eötvös Loránd Research Network, Temesvári krt. 62, Szeged, 6726 Hungary ,Single-Cell Technologies Ltd, Temesvári krt. 62, Szeged, 6726 Hungary
| | - Péter Horváth
- grid.481814.00000 0004 0479 9817Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Eötvös Loránd Research Network, Temesvári krt. 62, Szeged, 6726 Hungary ,Single-Cell Technologies Ltd, Temesvári krt. 62, Szeged, 6726 Hungary ,grid.7737.40000 0004 0410 2071Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014 Helsinki, Finland
| | - Bence Kővári
- grid.9008.10000 0001 1016 9625Department of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, Állomás utca 1, Szeged, 6720 Hungary
| | - Gábor Cserni
- grid.9008.10000 0001 1016 9625Department of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, Állomás utca 1, Szeged, 6720 Hungary
| | - Tamás Csont
- grid.9008.10000 0001 1016 9625MEDICS Research Group, Department of Biochemistry, University of Szeged Albert Szent-Györgyi Medical School, Dóm tér 9, Szeged, 6720 Hungary ,grid.9008.10000 0001 1016 9625Interdisciplinary Center of Excellence, University of Szeged, Dugonics tér 13, Szeged, 6720 Hungary
| | - Miklós Sántha
- grid.481814.00000 0004 0479 9817Laboratory of Animal Genetics and Molecular Neurobiology, Institute of Biochemistry, Biological Research Centre, Eötvös Loránd Research Network, Temesvári krt. 62, Szeged, 6726 Hungary
| |
Collapse
|
7
|
Lin Y, Peng X, Lin X, Deng X, Liu F, Tao H, Dong R, Wang B, Bi Y. Potential Value of Serum Lipid in the Identication of Postoperative Delirium Undergoing Knee/Hip Arthroplasty: The Perioperative Neurocognitive Disorder and Biomarker Lifestyle Study. Front Psychiatry 2022; 13:870317. [PMID: 35492710 PMCID: PMC9039337 DOI: 10.3389/fpsyt.2022.870317] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 03/18/2022] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE We aimed to investigate the relationship between preoperative lipid level and postoperative delirium (POD) and explore whether lipid's effect on POD is mediated by POD core protein. METHODS A total of 635 patients who were planned to undergo knee/hip arthroplasty under combined spinal-epidural anesthesia, regardless of gender, were selected. The patients were aged 40-90 years with American Society of Anesthesiologists physical status I II. The Mini-Mental State Examination (MMSE) was completed 1 day before the operation. Five milliliter elbow venous blood was taken from the patients before anesthesia, and serum levels of total cholesterol (TG), triglyceride (TC), low-density lipoprotein (LDL-C), and high-density lipoprotein (HDL-C) were detected. Cerebrospinal fluid (CSF) was extracted after successful spinal-epidural combined puncture, and amyloid beta40 (Aβ40), amyloid beta42 (Aβ42), total Tau (t-Tau), and phosphorylated Tau (p-Tau) in the CSF were measured by enzyme-linked immunosorbent assays (ELISA). After the operation, the occurrence and severity of POD were assessed using the Confusion Assessment Method and the Memorial Delirium Assessment Scale (MDAS), respectively. Patients were categorized into POD group and NPOD group. Logistic regression was used to analyze the relationship between POD and TC, TG, LDL-C, and HDL-C, and the mediating effect was used to analyze the role of POD core proteins in the relationship between lipid and MDAS. We used the receiver operating characteristic (ROC) and the precision-recall curve (PRC) analysis to assess the ability of TC, TG, LDL-C, and HDL-C ability to predict POD. Finally, we performed a sensitivity analysis to assess the stability of the results. RESULTS A total of 562 patients were finally enrolled in this study, and 66 patients developed POD, with an incidence of 11.7%. Logistic regression analysis showed that high concentration of TC (OR = 3.148, 95%CI 1.858∼5.333, P < 0.001), TG (OR = 2.483, 95%CI 1.573∼3.918, P < 0.001), and LDL-C (OR = 2.469, 95%CI 1.310∼4.656, P = 0.005) in serum were risk factors for POD. A high concentration of HDL-C (OR = 0.258, 95%CI 0.112∼0.594, P = 0.001) was a protective factor for POD after adjusted for age, sex, education, and MMSE score. ROC curves showed that HDL-C have the highest sensitivity and specificity in predicting POD. For these four lipid markers, the PRC range from 0.602 to 0.731, respectively. The mediating analysis showed that POD core proteins could partially mediate the relationship between lipid and POD (effect value: 16.19∼91.04%). The results were barely changed in the sensitivity analysis, and the sensitivity analysis has shown that the results were stable. CONCLUSION The increase of serum TG, TC, and LDL-C concentration is a risk factor for POD development, while high HDL-C concentration is a protective factor for POD, and the occurrence of POD is caused by hyperlipidemia may be caused by POD core proteins. CLINICAL TRIAL REGISTRATION [www.ClinicalTrials.gov], identifier [Chictr200033439].
Collapse
Affiliation(s)
- Yanan Lin
- Department of Anesthesiology, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, China
| | - Xiaoyan Peng
- Department of Anesthesiology, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, China
| | - Xu Lin
- Department of Anesthesiology, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, China
| | - Xiyuan Deng
- Department of Anesthesiology, Dalian Medical University, Dalian, China
| | - Fanghao Liu
- Department of Anesthesiology, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, China
| | - He Tao
- Department of Anesthesiology, Dalian Medical University, Dalian, China
| | - Rui Dong
- Department of Anesthesiology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, China
| | - Bin Wang
- Department of Anesthesiology, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, China
| | - Yanlin Bi
- Department of Anesthesiology, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, China
| |
Collapse
|
8
|
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: 23] [Impact Index Per Article: 7.7] [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
|
9
|
Varga-Medveczky Z, Kovács N, Tóth ME, Sántha M, Horváth I, Bors LA, Fónagy K, Imre T, Szabó P, Máthé D, Erdő F. Age-Related Inflammatory Balance Shift, Nasal Barrier Function, and Cerebro-Morphological Status in Healthy and Diseased Rodents. Front Neurosci 2021; 15:700729. [PMID: 34366780 PMCID: PMC8343234 DOI: 10.3389/fnins.2021.700729] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 06/14/2021] [Indexed: 01/20/2023] Open
Abstract
Increased blood–brain barrier (BBB) permeability and extensive neuronal changes have been described earlier in both healthy and pathological aging like apolipoprotein B-100 (APOB-100) and amyloid precursor protein (APP)–presenilin-1 (PSEN1) transgenic mouse models. APOB-100 hypertriglyceridemic model is a useful tool to study the link between cerebrovascular pathology and neurodegeneration, while APP–PSEN1 humanized mouse is a model of Alzheimer’s disease. The aim of the current study was to characterize the inflammatory changes in the brain with healthy aging and in neurodegeneration. Also, the cerebro-morphological and cognitive alterations have been investigated. The nose-to-brain delivery of a P-glycoprotein substrate model drug (quinidine) was monitored in the disease models and compared with the age-matched controls. Our results revealed an inflammatory balance shift in both the healthy aged and neurodegenerative models. In normal aging monocyte chemoattractant protein-1, stem cell factor and Rantes were highly upregulated indicating a stimulated leukocyte status. In APOB-100 mice, vascular endothelial growth factor (VEGF), platelet-derived growth factor (PDGF-BB), and interleukin-17A (IL-17A) were induced (vascular reaction), while in APP–PSEN1 mice resistin, IL-17A and GM-CSF were mostly upregulated. The nasal drug absorption was similar in the brain and blood indicating the molecular bypass of the BBB. The learning and memory tests showed no difference in the cognitive performance of healthy aged and young animals. Based on these results, it can be concluded that various markers of chronic inflammation are present in healthy aged and diseased animals. In APOB-100 mice, a cerebro-ventricular dilation can also be observed. For development of proper anti-aging and neuroprotective compounds, further studies focusing on the above inflammatory targets are suggested.
Collapse
Affiliation(s)
- Zsófia Varga-Medveczky
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - Noémi Kovács
- Department of Biophysics and Radiation Biology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Melinda E Tóth
- Institute of Biochemistry, ELKH Biological Research Centre, Szeged, Hungary
| | - Miklós Sántha
- Institute of Biochemistry, ELKH Biological Research Centre, Szeged, Hungary
| | - Ildikó Horváth
- Department of Biophysics and Radiation Biology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Luca Anna Bors
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary.,Heart and Vascular Centre, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Katalin Fónagy
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - Timea Imre
- Research Centre for Natural Sciences, Centre for Structural Study, Budapest, Hungary
| | - Pál Szabó
- Research Centre for Natural Sciences, Centre for Structural Study, Budapest, Hungary
| | - Domokos Máthé
- Department of Biophysics and Radiation Biology, Faculty of Medicine, Semmelweis University, Budapest, Hungary.,Hungarian Center of Excellence for Molecular Medicine (HCEMM), Advanced In Vivo Imaging Core Faciltiy, Budapest, Hungary
| | - Franciska Erdő
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| |
Collapse
|
10
|
Fonar G, Polis B, Sams DS, Levi A, Malka A, Bal N, Maltsev A, Elliott E, Samson AO. Modified Snake α-Neurotoxin Averts β-Amyloid Binding to α7 Nicotinic Acetylcholine Receptor and Reverses Cognitive Deficits in Alzheimer's Disease Mice. Mol Neurobiol 2021; 58:2322-2341. [PMID: 33417228 PMCID: PMC8018932 DOI: 10.1007/s12035-020-02270-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 12/18/2020] [Indexed: 12/03/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of senile dementia and one of the greatest medical, social, and economic challenges. According to a dominant theory, amyloid-β (Aβ) peptide is a key AD pathogenic factor. Aβ-soluble species interfere with synaptic functions, aggregate gradually, form plaques, and trigger neurodegeneration. The AD-associated pathology affects numerous systems, though the substantial loss of cholinergic neurons and α7 nicotinic receptors (α7AChR) is critical for the gradual cognitive decline. Aβ binds to α7AChR under various experimental settings; nevertheless, the functional significance of this interaction is ambiguous. Whereas the capability of low Aβ concentrations to activate α7AChR is functionally beneficial, extensive brain exposure to high Aβ concentrations diminishes α7AChR activity, contributes to the cholinergic deficits that characterize AD. Aβ and snake α-neurotoxins competitively bind to α7AChR. Accordingly, we designed a chemically modified α-cobratoxin (mToxin) to inhibit the interaction between Aβ and α7AChR. Subsequently, we examined mToxin in a set of original in silico, in vitro, ex vivo experiments, and in a murine AD model. We report that mToxin reversibly inhibits α7AChR, though it attenuates Aβ-induced synaptic transmission abnormalities, and upregulates pathways supporting long-term potentiation and reducing apoptosis. Remarkably, mToxin demonstrates no toxicity in brain slices and mice. Moreover, its chronic intracerebroventricular administration improves memory in AD-model animals. Our results point to unique mToxin neuroprotective properties, which might be tailored for the treatment of AD. Our methodology bridges the gaps in understanding Aβ-α7AChR interaction and represents a promising direction for further investigations and clinical development.
Collapse
Affiliation(s)
- Gennadiy Fonar
- Drug Discovery Laboratory, The Azrieli Faculty of Medicine, Bar-Ilan University, 1311502, Safed, Israel.
| | - Baruh Polis
- Drug Discovery Laboratory, The Azrieli Faculty of Medicine, Bar-Ilan University, 1311502, Safed, Israel
| | - Dev Sharan Sams
- Laboratory of Molecular and Behavioral Neuroscience, The Azrieli Faculty of Medicine, Bar-Ilan University, 1311502, Safed, Israel
| | - Almog Levi
- Drug Discovery Laboratory, The Azrieli Faculty of Medicine, Bar-Ilan University, 1311502, Safed, Israel
| | - Assaf Malka
- Drug Discovery Laboratory, The Azrieli Faculty of Medicine, Bar-Ilan University, 1311502, Safed, Israel
| | - Natalia Bal
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, Russia
| | - Alexander Maltsev
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, Russia
| | - Evan Elliott
- Laboratory of Molecular and Behavioral Neuroscience, The Azrieli Faculty of Medicine, Bar-Ilan University, 1311502, Safed, Israel
| | - Abraham O Samson
- Drug Discovery Laboratory, The Azrieli Faculty of Medicine, Bar-Ilan University, 1311502, Safed, Israel
| |
Collapse
|
11
|
Tóth ME, Dukay B, Péter M, Balogh G, Szűcs G, Zvara Á, Szebeni GJ, Hajdu P, Sárközy M, Puskás LG, Török Z, Csont T, Vígh L, Sántha M. Male and Female Animals Respond Differently to High-Fat Diet and Regular Exercise Training in a Mouse Model of Hyperlipidemia. Int J Mol Sci 2021; 22:ijms22084198. [PMID: 33919597 PMCID: PMC8073713 DOI: 10.3390/ijms22084198] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/13/2021] [Accepted: 04/15/2021] [Indexed: 01/18/2023] Open
Abstract
Inappropriate nutrition and a sedentary lifestyle can lead to obesity, one of the most common risk factors for several chronic diseases. Although regular physical exercise is an efficient approach to improve cardiometabolic health, the exact cellular processes are still not fully understood. We aimed to analyze the morphological, gene expression, and lipidomic patterns in the liver and adipose tissues in response to regular exercise. Healthy (wild type on a normal diet) and hyperlipidemic, high-fat diet-fed (HFD-fed) apolipoprotein B-100 (APOB-100)-overexpressing mice were trained by treadmill running for 7 months. The serum concentrations of triglyceride and tumor necrosis factor α (TNFα), as well as the level of lipid accumulation in the liver, were significantly higher in HFD-fed APOB-100 males compared to females. However, regular exercise almost completely abolished lipid accumulation in the liver of hyperlipidemic animals. The expression level of the thermogenesis marker, uncoupling protein-1 (Ucp1), was significantly higher in the subcutaneous white adipose tissue of healthy females, as well as in the brown adipose tissue of HFD-fed APOB-100 females, compared to males. Lipidomic analyses revealed that hyperlipidemia essentially remodeled the lipidome of brown adipose tissue, affecting both the membrane and storage lipid fractions, which was partially restored by exercise in both sexes. Our results revealed more severe metabolic disturbances in HFD-fed APOB-100 males compared to females. However, exercise efficiently reduced the body weight, serum triglyceride levels, expression of pro-inflammatory factors, and hepatic lipid accumulation in our model.
Collapse
Affiliation(s)
- Melinda E. Tóth
- Institute of Biochemistry, ELKH Biological Research Centre, H-6726 Szeged, Hungary; (B.D.); (M.P.); (G.B.); (P.H.); (Z.T.); (L.V.); (M.S.)
- Correspondence: ; Tel.: +36-62-599-635
| | - Brigitta Dukay
- Institute of Biochemistry, ELKH Biological Research Centre, H-6726 Szeged, Hungary; (B.D.); (M.P.); (G.B.); (P.H.); (Z.T.); (L.V.); (M.S.)
- Doctoral School in Biology, University of Szeged, H-6726 Szeged, Hungary
| | - Mária Péter
- Institute of Biochemistry, ELKH Biological Research Centre, H-6726 Szeged, Hungary; (B.D.); (M.P.); (G.B.); (P.H.); (Z.T.); (L.V.); (M.S.)
| | - Gábor Balogh
- Institute of Biochemistry, ELKH Biological Research Centre, H-6726 Szeged, Hungary; (B.D.); (M.P.); (G.B.); (P.H.); (Z.T.); (L.V.); (M.S.)
| | - Gergő Szűcs
- MEDICS Research Group, Department of Biochemistry, Interdisciplinary Center of Excellence, University of Szeged, H-6720 Szeged, Hungary; (G.S.); (M.S.); (T.C.)
| | - Ágnes Zvara
- Laboratory of Functional Genomics, ELKH Biological Research Centre, H-6726 Szeged, Hungary; (Á.Z.); (G.J.S.); (L.G.P.)
| | - Gábor J. Szebeni
- Laboratory of Functional Genomics, ELKH Biological Research Centre, H-6726 Szeged, Hungary; (Á.Z.); (G.J.S.); (L.G.P.)
| | - Petra Hajdu
- Institute of Biochemistry, ELKH Biological Research Centre, H-6726 Szeged, Hungary; (B.D.); (M.P.); (G.B.); (P.H.); (Z.T.); (L.V.); (M.S.)
| | - Márta Sárközy
- MEDICS Research Group, Department of Biochemistry, Interdisciplinary Center of Excellence, University of Szeged, H-6720 Szeged, Hungary; (G.S.); (M.S.); (T.C.)
| | - László G. Puskás
- Laboratory of Functional Genomics, ELKH Biological Research Centre, H-6726 Szeged, Hungary; (Á.Z.); (G.J.S.); (L.G.P.)
| | - Zsolt Török
- Institute of Biochemistry, ELKH Biological Research Centre, H-6726 Szeged, Hungary; (B.D.); (M.P.); (G.B.); (P.H.); (Z.T.); (L.V.); (M.S.)
| | - Tamás Csont
- MEDICS Research Group, Department of Biochemistry, Interdisciplinary Center of Excellence, University of Szeged, H-6720 Szeged, Hungary; (G.S.); (M.S.); (T.C.)
| | - László Vígh
- Institute of Biochemistry, ELKH Biological Research Centre, H-6726 Szeged, Hungary; (B.D.); (M.P.); (G.B.); (P.H.); (Z.T.); (L.V.); (M.S.)
| | - Miklós Sántha
- Institute of Biochemistry, ELKH Biological Research Centre, H-6726 Szeged, Hungary; (B.D.); (M.P.); (G.B.); (P.H.); (Z.T.); (L.V.); (M.S.)
| |
Collapse
|
12
|
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: 2.5] [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
|
13
|
Jiang S, Zhang CY, Tang L, Zhao LX, Chen HZ, Qiu Y. Integrated Genomic Analysis Revealed Associated Genes for Alzheimer's Disease in APOE4 Non-Carriers. Curr Alzheimer Res 2020; 16:753-763. [PMID: 31441725 DOI: 10.2174/1567205016666190823124724] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 07/14/2019] [Accepted: 08/08/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND APOE4 is the strongest genetic risk factor for late-onset Alzheimer's disease (LOAD). LOAD patients carrying or not carrying APOE4 manifest distinct clinico-pathological characteristics. APOE4 has been shown to play a critical role in the pathogenesis of AD by affecting various aspects of pathological processes. However, the pathogenesis involved in LOAD not-carrying APOE4 remains elusive. OBJECTIVE We aimed to identify the associated genes involved in LOAD not-carrying APOE4. METHODS An integrated genomic analysis of datasets of genome-wide association study, genome-wide expression profiling and genome-wide linkage scan and protein-protein interaction network construction were applied to identify associated gene clusters in APOE4 non-carriers. The role of one of hub gene of an APOE4 non-carrier-associated gene cluster in tau phosphorylation was studied by knockdown and western blot. RESULTS We identified 12 gene clusters associated with AD APOE4 non-carriers. The hub genes associated with AD in these clusters were MAPK8, POU2F1, XRCC1, PRKCG, EXOC6, VAMP4, SIRT1, MME, NOS1, ABCA1 and LDLR. The associated genes for APOE4 non-carriers were enriched in hereditary disorder, neurological disease and psychological disorders. Moreover, knockdown of PRKCG to reduce the expression of protein kinase Cγ isoform enhanced tau phosphorylation at Thr181 and Thr231 and the expression of glycogen synthase kinase 3β and cyclin-dependent kinase 5 in the presence of APOE3 but not APOE4. CONCLUSION The study provides new insight into the mechanism of distinct pathogenesis of LOAD not carrying APOE4 and prompts the functional exploration of identified genes based on APOE genotypes.
Collapse
Affiliation(s)
- Shan Jiang
- Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.,Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, United States
| | - Chun-Yun Zhang
- Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ling Tang
- Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Lan-Xue Zhao
- Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Hong-Zhuan Chen
- Institute of Interdisciplinary Integrative Biomedical Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201210, China
| | - Yu Qiu
- Department of Pharmacology and Chemical Biology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
14
|
Neuroprotection by Heat Shock Factor-1 (HSF1) and Trimerization-Deficient Mutant Identifies Novel Alterations in Gene Expression. Sci Rep 2018; 8:17255. [PMID: 30467350 PMCID: PMC6250741 DOI: 10.1038/s41598-018-35610-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 11/05/2018] [Indexed: 12/17/2022] Open
Abstract
Heat shock factor-1 (HSF1) protects neurons from death caused by the accumulation of misfolded proteins by stimulating the transcription of genes encoding heat shock proteins (HSPs). This stimulatory action depends on the association of trimeric HSF1 to sequences within HSP gene promoters. However, we recently described that HSF-AB, a mutant form of HSF1 that is incapable of either homo-trimerization, association with HSP gene promoters, or stimulation of HSP expression, protects neurons just as efficiently as wild-type HSF1 suggesting an alternative neuroprotective mechanism that is activated by HSF1. To gain insight into the mechanism by which HSF1 and HSF1-AB protect neurons, we used RNA-Seq technology to identify transcriptional alterations induced by these proteins in either healthy cerebellar granule neurons (CGNs) or neurons primed to die. When HSF1 was ectopically-expressed in healthy neurons, 1,211 differentially expressed genes (DEGs) were identified with 1,075 being upregulated. When HSF1 was expressed in neurons primed to die, 393 genes were upregulated and 32 genes were downregulated. In sharp contrast, HSF1-AB altered expression of 13 genes in healthy neurons and only 6 genes in neurons under apoptotic conditions, suggesting that the neuroprotective effect of HSF1-AB may be mediated by a non-transcriptional mechanism. We validated the altered expression of 15 genes by QPCR. Although other studies have conducted RNA-Seq analyses to identify HSF1 targets, our study performed using primary neurons has identified a number of novel targets that may play a special role in brain maintenance and function.
Collapse
|
15
|
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: 1.7] [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
|
16
|
Liu XJ, Wei J, Shang YH, Huang HC, Lao FX. Modulation of AβPP and GSK3β by Endoplasmic Reticulum Stress and Involvement in Alzheimer's Disease. J Alzheimers Dis 2018; 57:1157-1170. [PMID: 28339396 DOI: 10.3233/jad-161111] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) is a dementia disease with neuronal loss and synaptic impairment. This impairment is caused, at least partly, by the generation of two main AD hallmarks, namely the hyperphosphorylated tau protein comprising neurofibrillary tangles and senile plaques containing amyloid-β (Aβ) peptides. The amyloid-β protein precursor (AβPP) and glycogen synthase kinase-3β (GSK3β) are two main proteins associated with AD and are closely correlated with these hallmarks. Recently, both of the proteins were reported to be modulated by endoplasmic reticulum stress (ERS) and are involved in the pathogenesis of AD. The mechanism of ERS plus the modulation of AβPP processing and GSK3β activity by ERS in AD are summarized and explored in this review.
Collapse
Affiliation(s)
- Xin-Jun Liu
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, P.R. China.,College of Arts and Science of Beijing Union University, Beijing, P.R. China
| | - Jun Wei
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, P.R. China.,College of Arts and Science of Beijing Union University, Beijing, P.R. China
| | - Ying-Hui Shang
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, P.R. China.,College of Arts and Science of Beijing Union University, Beijing, P.R. China
| | - Han-Chang Huang
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, P.R. China.,College of Arts and Science of Beijing Union University, Beijing, P.R. China
| | - Feng-Xue Lao
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, P.R. China.,College of Arts and Science of Beijing Union University, Beijing, P.R. China
| |
Collapse
|
17
|
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: 2.9] [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
|
18
|
Low dosage of rimonabant leads to anxiolytic-like behavior via inhibiting expression levels and G-protein activity of kappa opioid receptors in a cannabinoid receptor independent manner. Neuropharmacology 2015; 89:298-307. [DOI: 10.1016/j.neuropharm.2014.10.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 09/19/2014] [Accepted: 10/04/2014] [Indexed: 12/15/2022]
|
19
|
Shared mechanisms of neurodegeneration in Alzheimer's disease and Parkinson's disease. BIOMED RESEARCH INTERNATIONAL 2014; 2014:648740. [PMID: 24900975 PMCID: PMC4037122 DOI: 10.1155/2014/648740] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2014] [Revised: 04/16/2014] [Accepted: 04/20/2014] [Indexed: 12/03/2022]
Abstract
Alzheimer's disease (AD) and Parkinson's disease (PD) have markedly different clinical and pathological features, but these two diseases are the most common neurodegenerative disorders. Previous studies have showed that there are common mechanisms in AD and PD. Several genetic studies have revealed mutations in genes associated with the risk of AD and PD. Circumstantial evidences have shown that dysregulation of brain iron homeostasis leads to abnormal iron accumulation and results in AD as well as PD. α-Synuclein and tau take part in the mechanisms of these diseases by oxidative stress and mitochondrial dysfunction. Some studies indicated that the loss of LC noradrenergic neurons may occur early in the progression of AD and PD. Nicotinic acetylcholine receptors (nAChRs) are members of the Cys-loop superfamily of pentameric ligand-gated ion channels; some evidence showed that nicotinic receptors may be associated with AD and PD. These experimental and clinical studies may provide a scientific foundation for common shared mechanisms in AD and PD.
Collapse
|
20
|
Abstract
Cellular and organismal aging are driven in part by the MTOR (mechanistic target of rapamycin) pathway and rapamycin extends life span in C elegans, Drosophila and mice. Herein, we investigated effects of rapamycin on brain aging in OXYS rats. Previously we found, in OXYS rats, an early development of age-associated pathological phenotypes similar to several geriatric disorders in humans, including cerebral dysfunctions. Behavioral alterations as well as learning and memory deficits develop by 3 months. Here we show that rapamycin treatment (0.1 or 0.5 mg/kg as a food mixture daily from the age of 1.5 to 3.5 months) decreased anxiety and improved locomotor and exploratory behavior in OXYS rats. In untreated OXYS rats, MRI revealed an increase of the area of hippocampus, substantial hydrocephalus and 2-fold increased area of the lateral ventricles. Rapamycin treatment prevented these abnormalities, erasing the difference between OXYS and Wistar rats (used as control). All untreated OXYS rats showed signs of neurodegeneration, manifested by loci of demyelination. Rapamycin decreased the percentage of animals with demyelination and the number of loci. Levels of Tau and phospho-Tau (T181) were increased in OXYS rats (compared with Wistar). Rapamycin significantly decreased Tau and inhibited its phosphorylation in the hippocampus of OXYS and Wistar rats. Importantly, rapamycin treatment caused a compensatory increase in levels of S6 and correspondingly levels of phospo-S6 in the frontal cortex, indicating that some downstream events were compensatory preserved, explaining the lack of toxicity. We conclude that rapamycin in low chronic doses can suppress brain aging.
Collapse
|
21
|
Impact of ApoB-100 expression on cognition and brain pathology in wild-type and hAPPsl mice. Neurobiol Aging 2013; 34:2379-88. [DOI: 10.1016/j.neurobiolaging.2013.04.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Revised: 03/12/2013] [Accepted: 04/03/2013] [Indexed: 11/22/2022]
|
22
|
Calderón-Garcidueñas L, Franco-Lira M, Mora-Tiscareño A, Medina-Cortina H, Torres-Jardón R, Kavanaugh M. Early Alzheimer's and Parkinson's disease pathology in urban children: Friend versus Foe responses--it is time to face the evidence. BIOMED RESEARCH INTERNATIONAL 2013; 2013:161687. [PMID: 23509683 PMCID: PMC3581281 DOI: 10.1155/2013/161687] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Revised: 01/01/2013] [Accepted: 01/01/2013] [Indexed: 01/28/2023]
Abstract
Chronic exposure to particulate matter air pollution is known to cause inflammation leading to respiratory- and cardiovascular-related sickness and death. Mexico City Metropolitan Area children exhibit an early brain imbalance in genes involved in oxidative stress, inflammation, and innate and adaptive immune responses. Early dysregulated neuroinflammation, brain microvascular damage, production of potent vasoconstrictors, and perturbations in the integrity of the neurovascular unit likely contribute to progressive neurodegenerative processes. The accumulation of misfolded proteins coincides with the anatomical distribution observed in the early stages of both Alzheimer's and Parkinson's diseases. We contend misfolding of hyperphosphorylated tau (HPπ), alpha-synuclein, and beta-amyloid could represent a compensatory early protective response to the sustained systemic and brain inflammation. However, we favor the view that the chronic systemic and brain dysregulated inflammation and the diffuse vascular damage contribute to the establishment of neurodegenerative processes with childhood clinical manifestations. Friend turns Foe early; therefore, implementation of neuroprotective measures to ameliorate or stop the inflammatory and neurodegenerative processes is warranted in exposed children. Epidemiological, cognitive, structural, and functional neuroimaging and mechanistic studies into the association between air pollution exposures and the development of neuroinflammation and neurodegeneration in children are of pressing importance for public health.
Collapse
Affiliation(s)
- Lilian Calderón-Garcidueñas
- Center for Structural and Functional Neurosciences, The University of Montana, 32 Campus Drive, Skaggs Building 287, Missoula, MT 59812, USA.
| | | | | | | | | | | |
Collapse
|