1
|
Darabi S, Gorgich EAC, Moradi F, Rustamzadeh A. Lipidopathy disrupts peripheral and central amyloid clearance in Alzheimer's disease: Where are our knowledge. IBRO Neurosci Rep 2025; 18:191-199. [PMID: 39906286 PMCID: PMC11791331 DOI: 10.1016/j.ibneur.2025.01.004] [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: 08/15/2024] [Revised: 12/09/2024] [Accepted: 01/04/2025] [Indexed: 02/06/2025] Open
Abstract
Amyloid-beta (Aβ) production is a normal physiological process, essential for neuronal function. However, an imbalance in Aβ production and clearance is the central pathological feature of Alzheimer's disease (AD), leading to the accumulation of Aβ plaques in the brain. Low-density lipoprotein receptor-related protein 1 (LRP1) plays a critical role in both the central clearance of Aβ from the brain and its peripheral transport to visceral organs. Disruptions in these processes contribute to the accumulation of Aβ in the central nervous system (CNS) and the progression of AD. Recent research emphasizes the need for a broader focus on the systemic effects of organs outside the brain, particularly in the context of AD prevention and treatment. The contribution of peripheral systems, such as the liver, in Aβ clearance, is vital, given that Aβ levels in the plasma correlate closely with those in the brain. Consequently, targeting systemic processes, rather than focusing solely on the CNS, may offer promising therapeutic approaches. Furthermore, high-density lipoprotein (HDL) facilitates the formation of lipoprotein-amyloid complexes, which are important for Aβ transport and clearance, using proteins such as apolipoproteins E and J (ApoE and ApoJ) to form complexes that help manage Aβ accumulation. On the other hand, low-density lipoprotein (LDL) facilitates Aβ efflux from the brain by binding to LRP1, promoting its clearance. Given the relationship between lipid profiles and Aβ levels, along with lipid-modifying drugs, may be effective in managing Aβ accumulation and mitigating AD progression.
Collapse
Affiliation(s)
- Shahram Darabi
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-communicable Diseases, Department of Anatomical Sciences, School of Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | | | - Fatemeh Moradi
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Auob Rustamzadeh
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-communicable Diseases, Department of Anatomical Sciences, School of Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| |
Collapse
|
2
|
Chen Y, Wang Z, Huang Q, Wang Y, Yan F, Xiang S, Xu L, Chen Y, Liu X, Chen G, Li M, Zhou Y. Differential proteomic profiles of exosomes in pediatric and adult adamantinomatous craniopharyngioma cyst fluid. Mol Biol Rep 2024; 51:1126. [PMID: 39505756 DOI: 10.1007/s11033-024-10073-y] [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: 08/14/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024]
Abstract
BACKGROUND Adamantinomatous craniopharyngiomas (ACPs), commonly seen in pediatrics and adults often present with large cystic cavities that can compress surrounding tissues, causing severe visual and endocrine symptoms. Complete resection of cystic ACP is challenging, frequently leading to postoperative recurrence. The composition of the cystic fluid is complex, and to date, there has been limited research focusing on exosomes within ACP cyst fluid. METHODS We collected cyst fluid from 12 ACP patients and confirmed the presence of exosomes. Subsequently, we conducted exosomal proteomic analysis using LC-MS/MS. The patients were divided into pediatric and adult groups for the analysis of differential protein enrichment, followed by comprehensive bioinformatics analysis, including GO analysis, KEGG analysis, and PPI network analysis, among other functional pathway and protein interaction analyses. Immunohistochemistry was used to determine the tissue expression distribution of the differential protein APOA1. RESULTS In our data analysis, 64 significantly differentially expressed proteins were identified, with 37 being overexpressed in the pediatric group and 27 in the adult group. Our results revealed that exosomal proteins in the pediatric group were predominantly enriched in modules and pathways related to high-density lipoprotein particle, apolipoprotein receptor binding, and the PPAR signaling pathway. Additionally, APOA1, as the hub protein with the highest connectivity in the differential protein interaction network, may play a critical role in β-amyloid metabolism pathways in pediatric ACP. CONCLUSION This study is the first to construct a proteomic map of ACP cyst fluid exosomes, suggesting significant differences in the tumor microenvironment's lipid metabolism between pediatrics and adults.
Collapse
Affiliation(s)
- Yiguang Chen
- Department of Neurosurgery, Xuanwu Hospital Capital Medical University, Beijing, 100053, China
| | - Ziyu Wang
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, 100053, China
| | - Qin Huang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou North Road, Guangzhou, 510515, Guangdong, China
| | - Yaming Wang
- Department of Neurosurgery, Xuanwu Hospital Capital Medical University, Beijing, 100053, China
| | - Feng Yan
- Department of Neurosurgery, Xuanwu Hospital Capital Medical University, Beijing, 100053, China
| | - Sishi Xiang
- Department of Neurosurgery, Xuanwu Hospital Capital Medical University, Beijing, 100053, China
| | - Lixin Xu
- Department of Neurosurgery, Xuanwu Hospital Capital Medical University, Beijing, 100053, China
| | - Yongjian Chen
- Dermatology and Venereology Division, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institute, 10005, Stockholm, Sweden.
| | - Xiaohai Liu
- Department of Neurosurgery, Xuanwu Hospital Capital Medical University, Beijing, 100053, China
| | - Ge Chen
- Department of Neurosurgery, Xuanwu Hospital Capital Medical University, Beijing, 100053, China
| | - Mingchu Li
- Department of Neurosurgery, Xuanwu Hospital Capital Medical University, Beijing, 100053, China
| | - Yiqiang Zhou
- Department of Neurosurgery, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.
| |
Collapse
|
3
|
Huang HX, Inglese P, Tang J, Yagoubi R, Correia GDS, Horneffer-van der Sluis VM, Camuzeaux S, Wu V, Kopanitsa MV, Willumsen N, Jackson JS, Barron AM, Saito T, Saido TC, Gentlemen S, Takats Z, Matthews PM. Mass spectrometry imaging highlights dynamic patterns of lipid co-expression with Aβ plaques in mouse and human brains. J Neurochem 2024; 168:1193-1214. [PMID: 38372586 DOI: 10.1111/jnc.16042] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 11/13/2023] [Accepted: 12/06/2023] [Indexed: 02/20/2024]
Abstract
Lipids play crucial roles in the susceptibility and brain cellular responses to Alzheimer's disease (AD) and are increasingly considered potential soluble biomarkers in cerebrospinal fluid (CSF) and plasma. To delineate the pathological correlations of distinct lipid species, we conducted a comprehensive characterization of both spatially localized and global differences in brain lipid composition in AppNL-G-F mice with spatial and bulk mass spectrometry lipidomic profiling, using human amyloid-expressing (h-Aβ) and WT mouse brains controls. We observed age-dependent increases in lysophospholipids, bis(monoacylglycerol) phosphates, and phosphatidylglycerols around Aβ plaques in AppNL-G-F mice. Immunohistology-based co-localization identified associations between focal pro-inflammatory lipids, glial activation, and autophagic flux disruption. Likewise, in human donors with varying Braak stages, similar studies of cortical sections revealed co-expression of lysophospholipids and ceramides around Aβ plaques in AD (Braak stage V/VI) but not in earlier Braak stage controls. Our findings in mice provide evidence of temporally and spatially heterogeneous differences in lipid composition as local and global Aβ-related pathologies evolve. Observing similar lipidomic changes associated with pathological Aβ plaques in human AD tissue provides a foundation for understanding differences in CSF lipids with reported clinical stage or disease severity.
Collapse
Affiliation(s)
- Helen Xuexia Huang
- Section of Bioanalytical Chemistry, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College London, Imperial College London, London, UK
| | - Paolo Inglese
- Section of Bioanalytical Chemistry, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- National Phenome Centre, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Jiabin Tang
- Department of Brain Sciences, Imperial College London, London, UK
| | - Riad Yagoubi
- Section of Bioanalytical Chemistry, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College London, Imperial College London, London, UK
| | - Gonçalo D S Correia
- Section of Bioanalytical Chemistry, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- National Phenome Centre, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | | | - Stephane Camuzeaux
- National Phenome Centre, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Vincen Wu
- Section of Bioanalytical Chemistry, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Maksym V Kopanitsa
- UK Dementia Research Institute at Imperial College London, Imperial College London, London, UK
| | - Nanet Willumsen
- UK Dementia Research Institute at Imperial College London, Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Johanna S Jackson
- UK Dementia Research Institute at Imperial College London, Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Anna M Barron
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Takashi Saito
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama, Japan
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University, Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama, Japan
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University, Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Steve Gentlemen
- Department of Brain Sciences, Imperial College London, London, UK
| | - Zoltan Takats
- Section of Bioanalytical Chemistry, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Paul M Matthews
- UK Dementia Research Institute at Imperial College London, Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| |
Collapse
|
4
|
Bonanni R, Cariati I, Cifelli P, Frank C, Annino G, Tancredi V, D'Arcangelo G. Exercise to Counteract Alzheimer's Disease: What Do Fluid Biomarkers Say? Int J Mol Sci 2024; 25:6951. [PMID: 39000060 PMCID: PMC11241657 DOI: 10.3390/ijms25136951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/14/2024] [Accepted: 06/22/2024] [Indexed: 07/16/2024] Open
Abstract
Neurodegenerative diseases (NDs) represent an unsolved problem to date with an ever-increasing population incidence. Particularly, Alzheimer's disease (AD) is the most widespread ND characterized by an accumulation of amyloid aggregates of beta-amyloid (Aβ) and Tau proteins that lead to neuronal death and subsequent cognitive decline. Although neuroimaging techniques are needed to diagnose AD, the investigation of biomarkers within body fluids could provide important information on neurodegeneration. Indeed, as there is no definitive solution for AD, the monitoring of these biomarkers is of strategic importance as they are useful for both diagnosing AD and assessing the progression of the neurodegenerative state. In this context, exercise is known to be an effective non-pharmacological management strategy for AD that can counteract cognitive decline and neurodegeneration. However, investigation of the concentration of fluid biomarkers in AD patients undergoing exercise protocols has led to unclear and often conflicting results, suggesting the need to clarify the role of exercise in modulating fluid biomarkers in AD. Therefore, this critical literature review aims to gather evidence on the main fluid biomarkers of AD and the modulatory effects of exercise to clarify the efficacy and usefulness of this non-pharmacological strategy in counteracting neurodegeneration in AD.
Collapse
Affiliation(s)
- Roberto Bonanni
- Department of Biomedicine and Prevention, "Tor Vergata" University of Rome, 00133 Rome, Italy
| | - Ida Cariati
- Department of Systems Medicine, "Tor Vergata" University of Rome, 00133 Rome, Italy
| | - Pierangelo Cifelli
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Claudio Frank
- UniCamillus-Saint Camillus International University of Health Sciences, 00131 Rome, Italy
| | - Giuseppe Annino
- Department of Systems Medicine, "Tor Vergata" University of Rome, 00133 Rome, Italy
- Centre of Space Bio-Medicine, "Tor Vergata" University of Rome, 00133 Rome, Italy
- Sports Engineering Laboratory, Department of Industrial Engineering, "Tor Vergata" University of Rome, 00133 Rome, Italy
| | - Virginia Tancredi
- Department of Systems Medicine, "Tor Vergata" University of Rome, 00133 Rome, Italy
- Centre of Space Bio-Medicine, "Tor Vergata" University of Rome, 00133 Rome, Italy
| | - Giovanna D'Arcangelo
- Department of Systems Medicine, "Tor Vergata" University of Rome, 00133 Rome, Italy
- Centre of Space Bio-Medicine, "Tor Vergata" University of Rome, 00133 Rome, Italy
| |
Collapse
|
5
|
Li N, Gao Y, Li LT, Hu YD, Ling L, Jia N, Chen YJ, Meng YN, Jiang Y. Development and validation of a nomogram predictive model for cognitive impairment in cerebral small vessel disease: a comprehensive retrospective analysis. Front Neurol 2024; 15:1373306. [PMID: 38952470 PMCID: PMC11215066 DOI: 10.3389/fneur.2024.1373306] [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: 01/25/2024] [Accepted: 06/06/2024] [Indexed: 07/03/2024] Open
Abstract
Background Cerebral small vessel disease (CSVD) is a common neurodegenerative condition in the elderly, closely associated with cognitive impairment. Early identification of individuals with CSVD who are at a higher risk of developing cognitive impairment is crucial for timely intervention and improving patient outcomes. Objective The aim of this study is to construct a predictive model utilizing LASSO regression and binary logistic regression, with the objective of precisely forecasting the risk of cognitive impairment in patients with CSVD. Methods The study utilized LASSO regression for feature selection and logistic regression for model construction in a cohort of CSVD patients. The model's validity was assessed through calibration curves and decision curve analysis (DCA). Results A nomogram was developed to predict cognitive impairment, incorporating hypertension, CSVD burden, apolipoprotein A1 (ApoA1) levels, and age. The model exhibited high accuracy with AUC values of 0.866 and 0.852 for the training and validation sets, respectively. Calibration curves confirmed the model's reliability, and DCA highlighted its clinical utility. The model's sensitivity and specificity were 75.3 and 79.7% for the training set, and 76.9 and 74.0% for the validation set. Conclusion This study successfully demonstrates the application of machine learning in developing a reliable predictive model for cognitive impairment in CSVD. The model's high accuracy and robust predictive capability provide a crucial tool for the early detection and intervention of cognitive impairment in patients with CSVD, potentially improving outcomes for this specific condition.
Collapse
Affiliation(s)
- Ning Li
- Department of Neurology, Affiliated Hospital of Hebei University, Baoding, China
| | - Yan Gao
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Li-tao Li
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| | - Ya-dong Hu
- Department of Neurology, Affiliated Hospital of Hebei University, Baoding, China
| | - Li Ling
- Department of Neurology, Affiliated Hospital of Hebei University, Baoding, China
| | - Nan Jia
- Department of Neurology, Affiliated Hospital of Hebei University, Baoding, China
| | - Ya-jing Chen
- Department of Neurology, Affiliated Hospital of Hebei University, Baoding, China
| | - Ya-nan Meng
- Department of Neurology, Affiliated Hospital of Hebei University, Baoding, China
| | - Ye Jiang
- Department of Neurology, Affiliated Hospital of Hebei University, Baoding, China
| |
Collapse
|
6
|
Feng Q, Lu Y, Zhang R, Li Y, Zhao Z, Zhou H. Identification of differentially expressed exosome proteins in serum as potential biomarkers for cognitive impairments in cerebral small vessel disease. Neurosci Lett 2024; 822:137631. [PMID: 38211879 DOI: 10.1016/j.neulet.2024.137631] [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: 08/29/2023] [Revised: 12/25/2023] [Accepted: 01/08/2024] [Indexed: 01/13/2024]
Abstract
BACKGROUND Cognitive impairment arising from cerebral small vessel disease (CSVD) represents a critical subtype of vascular cognitive impairments (VCI) and is the primary cause of vascular dementia. However, identifying reliable clinical and laboratory indicators for this disease remain elusive. We hypothesize that plasma exosome proteins hold the potential to serve as biomarkers for the onset of cognitive dysfunction associated with cerebrovascular diseases. METHODS We employed TMT-based proteomics to discern variations in serum exosome proteomes between individuals with cognitive impairments due to CSVD and healthy volunteers. RESULTS Each group comprised 18 subjects, and through differential expression analysis, we identified 22 down-regulated and 8 up-regulated proteins between the two groups. Our research revealed 30 differentially expressed plasma exosome proteins, including histone, proteasome, clusterin and coagulation factor XIII, in individuals with cognitive impairments caused by CSVD. CONCLUSION The 30 differentially expressed plasma exosome proteins identified in our study are promising as biomarkers for diagnosing cognitive impairments resulting from CSVD. These findings may help us better understand the underlying pathological mechanisms involved in the diseases.
Collapse
Affiliation(s)
- Qian Feng
- Department of Neurology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Yanjing Lu
- Department of Neurology, Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Ruyang Zhang
- Department of Neurology, Suzhou Wuzhong People's Hospital, Suzhou, China
| | - Yifan Li
- Department of Neurology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Zhong Zhao
- Department of Neurology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China.
| | - Hua Zhou
- Department of Neurology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China.
| |
Collapse
|
7
|
Ma L, Yuan YX, Cheng FJ, Liu Y, Wei Q, Peng YF, Wang Y. The association between blood lipids and cognitive impairment in type 2 diabetes mellitus. Eur J Med Res 2024; 29:1. [PMID: 38167163 PMCID: PMC10763275 DOI: 10.1186/s40001-023-01574-w] [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: 08/16/2023] [Accepted: 12/07/2023] [Indexed: 01/05/2024] Open
Abstract
OBJECTIVE The study was performed to explore the association between blood lipids and cognitive impairment in patients with type 2 diabetes mellitus (T2DM). METHODS This study included 336 patients with T2DM. Relevant clinical data including total cholesterol (TC), high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), triglyceride (TG), apolipoprotein A1, apolipoprotein B were collected, and the Mini-Mental State Examination (MMSE) score and Montreal Cognitive Assessment (MoCA) score were used to assess the cognitive function in patients with T2DM. RESULTS Serum apolipoprotein A1 levels were significantly increased in T2DM patients with cognitive impairment compared with T2DM patients without cognitive impairment (p = 0.017). Serum apolipoprotein A1 levels were significantly negatively correlated with MoCA score (r = - 0.143, p = 0.009) and MMSE score (r = - 0.132, p = 0.016) in patients with T2DM. In multivariable-adjusted regression model, serum apolipoprotein A1 was independently associated with cognitive impairment in patients with T2DM (OR = 5.201, p = 0.024). CONCLUSION Serum apolipoprotein A1 is associated with cognitive impairment in patients with T2DM, but not TC, TG, HDL-C, LDL-C, and apolipoprotein B, indicating that increased serum apolipoprotein A1 may be a risk factor of cognitive impairment in patients with T2DM.
Collapse
Affiliation(s)
- Li Ma
- Department of Rehabilitative Medicine, Affiliated Zhongda Hospital of Southeast University, Nanjing, 210009, Jiangsu, China
| | - Yue-Xing Yuan
- Department of Endocrinology and Metabolism, Affiliated Zhongda Hospital of Southeast University, Nanjing, 210009, Jiangsu, China
| | - Feng-Jin Cheng
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210009, Jiangsu, China
| | - Yan Liu
- Zhongda Hospital, Institute of Diabetes, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Qiong Wei
- Department of Endocrinology and Metabolism, Affiliated Zhongda Hospital of Southeast University, Nanjing, 210009, Jiangsu, China.
| | - You-Fan Peng
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China.
| | - Yao Wang
- Department of Endocrinology and Metabolism, Affiliated Zhongda Hospital of Southeast University, Nanjing, 210009, Jiangsu, China.
| |
Collapse
|
8
|
Wang J, Xu L, Chen X, Wu J, Chen Y, Feng Z, Dong L, Yao D, Cai Q, Jian W, Li H, Duan M, Wang Z. Correlation Analysis of ApoB, ApoA1, and ApoB/ApoA1 with Cortical Morphology in Patients with Memory Complaints. J Alzheimers Dis 2024; 101:1137-1150. [PMID: 39302359 DOI: 10.3233/jad-230863] [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] [Indexed: 09/22/2024]
Abstract
Background Apolipoproteins and cortical morphology are closely associated with memory complaints, and both may contribute to the development of Alzheimer's disease. Objective To examine whether apolipoprotein B (ApoB), apolipoprotein A-1 (ApoA1), and their ratio (ApoB/ApoA1) are associated with cortical morphology in patients with memory complaints. Methods Ninety-seven patients underwent neuropsychological testing, measurements of ApoB, ApoA1, ApoB/ApoA1, plasma Alzheimer's biomarker, apolipoprotein E (ApoE) genotyping, and 3T structural magnetic resonance imaging (sMRI) scans. Based on sMRI scanning locations, patients were categorized into the University of Electronic Science and Technology (UESTC) and the Fourth People's Hospital of Chengdu (FPHC). The Computational Anatomy Toolbox within Statistical Parametric Mapping was used to calculate each patient's cortical morphology index based on sMRI data. The cortical morphology index and apolipoproteins were also analyzed. Results Significant positive correlations were found between ApoB and sulcal depth in the lateral occipital cortex among the UESTC, the FPHC, and the total sample groups, and negative correlations were observed between sulcal depth in the lateral occipital cortex and the scores of the Shape Trails Test Part A and B. In the FPHC group, the scores of the Montreal Cognitive Assessment Basic, delayed recall of the Auditory Verbal Learning Test, Animal Fluency Test and Boston Naming Test were positively correlated with the sulcal depth. Conclusions ApoB is associated with the sulcal depth in the lateral occipital cortex, potentially relating to speed/executive function in individuals with memory complaints.
Collapse
Affiliation(s)
- Jiayu Wang
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
- Department of Geriatrics, the Fourth People's Hospital of Chengdu, Chengdu, China
- Nursing School of Zunyi Medical University, Zunyi, China
| | - Lisi Xu
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
- Department of Geriatrics, the Fourth People's Hospital of Chengdu, Chengdu, China
| | - Xuemei Chen
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
- Department of Geriatrics, the Fourth People's Hospital of Chengdu, Chengdu, China
| | - Jiajing Wu
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
- Department of Geriatrics, the Fourth People's Hospital of Chengdu, Chengdu, China
- Nursing School of Zunyi Medical University, Zunyi, China
| | - Yu Chen
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
- Department of Radiology, the Fourth People's Hospital of Chengdu, Chengdu, China
| | - Ziqian Feng
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
- Department of Geriatrics, the Fourth People's Hospital of Chengdu, Chengdu, China
- Nursing School of Zunyi Medical University, Zunyi, China
| | - Li Dong
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
- Research Unit of NeuroInformation, Chinese Academy of Medical Sciences, Chengdu, China
- Sichuan Institute for Brain Science and Brain-Inspired Intelligence, Chengdu, China
| | - Dezhong Yao
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
- Research Unit of NeuroInformation, Chinese Academy of Medical Sciences, Chengdu, China
- Sichuan Institute for Brain Science and Brain-Inspired Intelligence, Chengdu, China
| | - Qingyan Cai
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
- Department of Geriatrics, the Fourth People's Hospital of Chengdu, Chengdu, China
| | - Wei Jian
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
- Department of Geriatrics, the Fourth People's Hospital of Chengdu, Chengdu, China
| | - Hongyi Li
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
- Department of Geriatrics, the Fourth People's Hospital of Chengdu, Chengdu, China
| | - MingJun Duan
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
- Department of Geriatrics, the Fourth People's Hospital of Chengdu, Chengdu, China
| | - Ziqi Wang
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Lab for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
- Department of Geriatrics, the Fourth People's Hospital of Chengdu, Chengdu, China
| |
Collapse
|
9
|
Sánchez-Ortí JV, Correa-Ghisays P, Balanzá-Martínez V, Selva-Vera G, Vila-Francés J, Magdalena-Benedito R, San-Martin C, Victor VM, Escribano-Lopez I, Hernandez-Mijares A, Vivas-Lalinde J, Crespo-Facorro B, Tabarés-Seisdedos R. Inflammation and lipid metabolism as potential biomarkers of memory impairment across type 2 diabetes mellitus and severe mental disorders. Prog Neuropsychopharmacol Biol Psychiatry 2023; 127:110817. [PMID: 37327846 DOI: 10.1016/j.pnpbp.2023.110817] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/20/2023] [Accepted: 06/12/2023] [Indexed: 06/18/2023]
Abstract
INTRODUCTION Neurocognitive impairment is a transdiagnostic feature across several psychiatric and cardiometabolic conditions. The relationship between inflammatory and lipid metabolism biomarkers and memory performance is not fully understood. This study aimed to identify peripheral biomarkers suitable to signal memory decline from a transdiagnostic and longitudinal perspective. METHODS Peripheral blood biomarkers of inflammation, oxidative stress and lipid metabolism were assessed twice over a 1-year period in 165 individuals, including 30 with schizophrenia (SZ), 42 with bipolar disorder (BD), 35 with major depressive disorder (MDD), 30 with type 2 diabetes mellitus (T2DM), and 28 healthy controls (HCs). Participants were stratified by memory performance quartiles, taking as a reference their global memory score (GMS) at baseline, into categories of high memory (H; n = 40), medium to high memory (MH; n = 43), medium to low memory (ML; n = 38) and low memory (L; n = 44). Exploratory and confirmatory factorial analysis, mixed one-way analysis of covariance and discriminatory analyses were performed. RESULTS L group was significantly associated with higher levels of tumor necrosis factor-alpha (TNF-α) and lower levels of apolipoprotein A1 (Apo-A1) compared to those from the MH and H groups (p < 0.05; η2p = 0.06-0.09), with small to moderate effect sizes. Moreover, the combination of interleukin-6 (IL-6), TNF-α, c-reactive protein (CRP), Apo-A1 and Apo-B compounded the transdiagnostic model that best discriminated between groups with different degrees of memory impairment (χ2 = 11.9-49.3, p < 0.05-0.0001). CONCLUSIONS Inflammation and lipid metabolism seem to be associated with memory across T2DM and severe mental illnesses (SMI). A panel of biomarkers may be a useful approach to identify individuals at greater risk of neurocognitive impairment. These findings may have a potential translational utility for early intervention and advance precision medicine in these disorders.
Collapse
Affiliation(s)
- Joan Vicent Sánchez-Ortí
- INCLIVA - Health Research Institute, Valencia, Spain; TMAP - Evaluation Unit in Personal Autonomy, Dependency and Serious Mental Disorders, University of Valencia, Valencia, Spain; Faculty of Psychology, University of Valencia, Valencia, Spain
| | - Patricia Correa-Ghisays
- INCLIVA - Health Research Institute, Valencia, Spain; Center for Biomedical Research in Mental Health Network (CIBERSAM), Health Institute, Carlos III, Madrid, Spain; TMAP - Evaluation Unit in Personal Autonomy, Dependency and Serious Mental Disorders, University of Valencia, Valencia, Spain; Faculty of Psychology, University of Valencia, Valencia, Spain; Teaching Unit of Psychiatry and Psychological Medicine, Department of Medicine, University of Valencia, Valencia, Spain.
| | - Vicent Balanzá-Martínez
- INCLIVA - Health Research Institute, Valencia, Spain; Center for Biomedical Research in Mental Health Network (CIBERSAM), Health Institute, Carlos III, Madrid, Spain; TMAP - Evaluation Unit in Personal Autonomy, Dependency and Serious Mental Disorders, University of Valencia, Valencia, Spain; Teaching Unit of Psychiatry and Psychological Medicine, Department of Medicine, University of Valencia, Valencia, Spain; Mental Health Unit of Catarroja, Valencia, Spain.
| | - Gabriel Selva-Vera
- INCLIVA - Health Research Institute, Valencia, Spain; Center for Biomedical Research in Mental Health Network (CIBERSAM), Health Institute, Carlos III, Madrid, Spain; TMAP - Evaluation Unit in Personal Autonomy, Dependency and Serious Mental Disorders, University of Valencia, Valencia, Spain; Teaching Unit of Psychiatry and Psychological Medicine, Department of Medicine, University of Valencia, Valencia, Spain
| | - Joan Vila-Francés
- IDAL - Intelligent Data Analysis Laboratory, University of Valencia, Valencia, Spain
| | | | - Constanza San-Martin
- Center for Biomedical Research in Mental Health Network (CIBERSAM), Health Institute, Carlos III, Madrid, Spain; TMAP - Evaluation Unit in Personal Autonomy, Dependency and Serious Mental Disorders, University of Valencia, Valencia, Spain; Department of Physiotherapy, University of Valencia, Valencia, Spain
| | - Víctor M Victor
- Service of Endocrinology and Nutrition, University Hospital Dr. Peset, Spain; Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), Valencia, Spain; Department of Physiology, University of Valencia, Valencia, Spain
| | | | | | | | - Benedicto Crespo-Facorro
- Center for Biomedical Research in Mental Health Network (CIBERSAM), Health Institute, Carlos III, Madrid, Spain; Department of Psychiatry, Faculty of Medicine, University of Sevilla, HU Virgen del Rocío IBIS, Spain
| | - Rafael Tabarés-Seisdedos
- INCLIVA - Health Research Institute, Valencia, Spain; Center for Biomedical Research in Mental Health Network (CIBERSAM), Health Institute, Carlos III, Madrid, Spain; TMAP - Evaluation Unit in Personal Autonomy, Dependency and Serious Mental Disorders, University of Valencia, Valencia, Spain; Teaching Unit of Psychiatry and Psychological Medicine, Department of Medicine, University of Valencia, Valencia, Spain
| |
Collapse
|
10
|
Pillai JA, Bena J, Bekris L, Kodur N, Kasumov T, Leverenz JB, Kashyap SR. Metabolic syndrome biomarkers relate to rate of cognitive decline in MCI and dementia stages of Alzheimer's disease. Alzheimers Res Ther 2023; 15:54. [PMID: 36927447 PMCID: PMC10018847 DOI: 10.1186/s13195-023-01203-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 03/07/2023] [Indexed: 03/18/2023]
Abstract
BACKGROUND The relationship between biomarkers of metabolic syndrome and insulin resistance, plasma triglyceride/HDL cholesterol (TG/HDL-C) ratio, on the rate of cognitive decline in mild cognitive impairment (MCI) and dementia stages of Alzheimer's disease (AD) is unknown. The role of peripheral and cerebrospinal fluid (CSF) levels of Apolipoprotein A1 (ApoA1), a key functional component of HDL, on cognitive decline also remains unclear among them. Here we evaluate baseline plasma TG/HDL-C ratio and CSF and plasma ApoA1 levels and their relation with cognitive decline in the MCI and Dementia stages of AD. PATIENTS AND METHODS A retrospective longitudinal study (156 participants; 106 MCI, 50 AD dementia) from the Alzheimer's Disease Neuroimaging Initiative, with an average of 4.0 (SD 2.8) years follow-up. Baseline plasma TG/HDL-C, plasma, and CSF ApoA1 and their relationship to inflammation and blood-brain barrier (BBB) biomarkers and longitudinal cognitive outcomes were evaluated. Multivariable linear mixed effect models were used to assess the effect of baseline analytes with longitudinal changes in Mini-Mental State Exam (MMSE), Clinical Dementia Rating-Sum of Boxes (CDR-SB), and Logical Memory delayed recall (LM) score after controlling for well-known covariates. RESULTS A total of 156 participants included 98 women, 63%; mean age was 74.9 (SD 7.3) years. At baseline, MCI and dementia groups did not differ significantly in TG/HDL-C (Wilcoxon W statistic = 0.39, p = 0.39) and CSF ApoA1 levels (W = 3642, p = 0.29), but the dementia group had higher plasma ApoA1 than the MCI group (W = 4615, p = 0.01). Higher TG/HDL-C ratio was associated with faster decline in CDR-SB among MCI and dementia groups. Higher plasma ApoA1 was associated with faster decline in MMSE and LM among MCI, while in contrast higher CSF ApoA1 levels related to slower cognitive decline in MMSE among MCI. CSF and plasma ApoA1 also show opposite directional correlations with biomarkers of BBB integrity. CSF but not plasma levels of ApoA1 positively correlated to inflammation analytes in the AGE-RAGE signaling pathway in diabetic complications (KEGG ID:KO04933). CONCLUSIONS Biomarkers of metabolic syndrome relate to rate of cognitive decline among MCI and dementia individuals. Elevated plasma TG/HDL-C ratio and plasma ApoA1 are associated with worse cognitive outcomes in MCI and dementia participants. CSF ApoA1 and plasma ApoA1 likely have different roles in AD progression in MCI stage.
Collapse
Affiliation(s)
- Jagan A Pillai
- Lou Ruvo Center for Brain Health, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, 9500 Euclid Ave/U10, Cleveland, OH, 44195, USA. .,Neurological Institute, Cleveland Clinic Lerner College of Medicine, Cleveland, OH, 44195, USA. .,Department of Neurology, Cleveland Clinic Lerner College of Medicine, Cleveland, OH, 44195, USA. .,Cleveland Clinic Lerner College of Medicine, Cleveland, OH, 44195, USA.
| | - James Bena
- Quantitative Health Sciences, Cleveland Clinic Lerner College of Medicine, Cleveland, OH, 44195, USA
| | - Lynn Bekris
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH, 44195, USA.,Lerner Research Institute, Cleveland Clinic Lerner College of Medicine, Cleveland, OH, 44195, USA
| | - Nandan Kodur
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH, 44195, USA
| | - Takhar Kasumov
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, 44272, USA
| | - James B Leverenz
- Lou Ruvo Center for Brain Health, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, 9500 Euclid Ave/U10, Cleveland, OH, 44195, USA.,Neurological Institute, Cleveland Clinic Lerner College of Medicine, Cleveland, OH, 44195, USA.,Department of Neurology, Cleveland Clinic Lerner College of Medicine, Cleveland, OH, 44195, USA.,Cleveland Clinic Lerner College of Medicine, Cleveland, OH, 44195, USA
| | - Sangeeta R Kashyap
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH, 44195, USA.,Division of Endocrinology, Diabetes and Metabolism, Weill Cornell Medicine New York Presbyterian, New York, NY, 10021, USA
| | | |
Collapse
|
11
|
Wu W, Shen A, Lee I, Miranda EG, Spratt H, Pappolla M, Fang X, Bao X. Changes of tRNA-Derived Fragments by Alzheimer's Disease in Cerebrospinal Fluid and Blood Serum. J Alzheimers Dis 2023; 96:1285-1304. [PMID: 37980659 PMCID: PMC10832608 DOI: 10.3233/jad-230412] [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] [Indexed: 11/21/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is the most common type of dementia, affecting individuals over 65. AD is also a multifactorial disease, with disease mechanisms incompletely characterized, and disease-modifying therapies are marginally effective. Biomarker signatures may shed light on the diagnosis, disease mechanisms, and the development of therapeutic targets. tRNA-derived RNA fragments (tRFs), a family of recently discovered small non-coding RNAs, have been found to be significantly enhanced in human AD hippocampus tissues. However, whether tRFs change in body fluids is unknown. OBJECTIVE To investigate whether tRFs in body fluids are impacted by AD. METHODS We first used T4 polynucleotide kinase-RNA-seq, a modified next-generation sequencing technique, to identify detectable tRFs in human cerebrospinal fluid and serum samples. The detectable tRFs were then compared in these fluids from control, AD, and mild cognitive impairment patients using tRF qRT-PCR. The stability of tRFs in serum was also investigated by checking the change in tRFs in response to protein digestion or exosome lysis. RESULTS Among various tRFs, tRF5-ProAGG seemed to be impacted by AD in both cerebrospinal fluid and serum. AD-impacted serum tRF5-ProAGG showed a correlation with the AD stage. Putative targets of tRF5-ProAGG in the hippocampus were also predicted by a computational algorithm, with some targets being validated experimentally and one of them being in a negative correlation with tRF5-ProAGG even using a small size of samples. CONCLUSIONS tRF5-ProAGG showed the potential as an AD biomarker and may play a role in disease progression.
Collapse
Affiliation(s)
- Wenzhe Wu
- Department of Pediatrics, The University of Texas Medical Branch, Galveston, TX, USA
| | - Audrey Shen
- Department of Human Physiology, Boston University, Boston, MA, USA
| | | | - Ernesto G. Miranda
- Department of Neurology and Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, Galveston, TX
| | - Heidi Spratt
- Department of Biostatistics and Data Science, The University of Texas Medical Branch, Galveston, TX
- The Institute of Translational Sciences, The University of Texas Medical Branch, Galveston, TX
| | - Miguel Pappolla
- Department of Neurology and Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, Galveston, TX
| | - Xiang Fang
- Department of Neurology and Mitchell Center for Neurodegenerative Diseases, The University of Texas Medical Branch, Galveston, TX
| | - Xiaoyong Bao
- Department of Pediatrics, The University of Texas Medical Branch, Galveston, TX, USA
- Department of Biostatistics and Data Science, The University of Texas Medical Branch, Galveston, TX
- The Institute of Translational Sciences, The University of Texas Medical Branch, Galveston, TX
- The Institute for Human Infections and Immunity, The University of Texas Medical Branch, Galveston, TX
| |
Collapse
|
12
|
Nassar A, Kodi T, Satarker S, Chowdari Gurram P, Upadhya D, SM F, Mudgal J, Nampoothiri M. Astrocytic MicroRNAs and Transcription Factors in Alzheimer's Disease and Therapeutic Interventions. Cells 2022; 11:cells11244111. [PMID: 36552875 PMCID: PMC9776935 DOI: 10.3390/cells11244111] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 12/13/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022] Open
Abstract
Astrocytes are important for maintaining cholesterol metabolism, glutamate uptake, and neurotransmission. Indeed, inflammatory processes and neurodegeneration contribute to the altered morphology, gene expression, and function of astrocytes. Astrocytes, in collaboration with numerous microRNAs, regulate brain cholesterol levels as well as glutamatergic and inflammatory signaling, all of which contribute to general brain homeostasis. Neural electrical activity, synaptic plasticity processes, learning, and memory are dependent on the astrocyte-neuron crosstalk. Here, we review the involvement of astrocytic microRNAs that potentially regulate cholesterol metabolism, glutamate uptake, and inflammation in Alzheimer's disease (AD). The interaction between astrocytic microRNAs and long non-coding RNA and transcription factors specific to astrocytes also contributes to the pathogenesis of AD. Thus, astrocytic microRNAs arise as a promising target, as AD conditions are a worldwide public health problem. This review examines novel therapeutic strategies to target astrocyte dysfunction in AD, such as lipid nanodiscs, engineered G protein-coupled receptors, extracellular vesicles, and nanoparticles.
Collapse
Affiliation(s)
- Ajmal Nassar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Triveni Kodi
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Sairaj Satarker
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Prasada Chowdari Gurram
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Dinesh Upadhya
- Centre for Molecular Neurosciences, Kasturba Medical College, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Fayaz SM
- Department of Biotechnology, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Jayesh Mudgal
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Madhavan Nampoothiri
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
- Correspondence:
| |
Collapse
|
13
|
Anusha-Kiran Y, Mol P, Dey G, Bhat FA, Chatterjee O, Deolankar SC, Philip M, Prasad TSK, Srinivas Bharath MM, Mahadevan A. Regional heterogeneity in mitochondrial function underlies region specific vulnerability in human brain ageing: Implications for neurodegeneration. Free Radic Biol Med 2022; 193:34-57. [PMID: 36195160 DOI: 10.1016/j.freeradbiomed.2022.09.027] [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: 08/02/2022] [Revised: 09/12/2022] [Accepted: 09/22/2022] [Indexed: 12/01/2022]
Abstract
Selective neuronal vulnerability (SNV) of specific neuroanatomical regions such as frontal cortex (FC) and hippocampus (HC) is characteristic of age-associated neurodegenerative diseases (NDDs), although its pathogenetic basis remains unresolved. We hypothesized that physiological differences in mitochondrial function in neuroanatomical regions could contribute to SNV. To investigate this, we evaluated mitochondrial function in human brains (age range:1-90 y) in FC, striatum (ST), HC, cerebellum (CB) and medulla oblongata (MD), using enzyme assays and quantitative proteomics. Striking differences were noted in resistant regions- MD and CB compared to the vulnerable regions- FC, HC and ST. At younger age (25 ± 5 y), higher activity of electron transport chain enzymes and upregulation of metabolic and antioxidant proteins were noted in MD compared to FC and HC, that was sustained with increasing age (≥65 y). In contrast, the expression of synaptic proteins was higher in FC, HC and ST (vs. MD). In line with this, quantitative phospho-proteomics revealed activation of upstream regulators (ERS, PPARα) of mitochondrial metabolism and inhibition of synaptic pathways in MD. Microtubule Associated Protein Tau (MAPT) showed overexpression in FC, HC and ST both in young and older age (vs. MD). MAPT hyperphosphorylation and the activation of its kinases were noted in FC and HC with age. Our study demonstrates that regional heterogeneity in mitochondrial and other cellular functions contribute to SNV and protect regions such as MD, while rendering FC and HC vulnerable to NDDs. The findings also support the "last in, first out" hypothesis of ageing, wherein regions such as FC, that are the most recent to develop phylogenetically and ontogenetically, are the first to be affected in ageing and NDDs.
Collapse
Affiliation(s)
- Yarlagadda Anusha-Kiran
- Department of Neuropathology, National Institute of Mental Health and Neurosciences (NIMHANS), No. 2900, Hosur Road, Bangalore, 560029, India; Department of Clinical Psychopharmacology and Neurotoxicology, NIMHANS, No. 2900, Hosur Road, Bangalore, 560029, India
| | - Praseeda Mol
- Institute of Bioinformatics, International Technology Park, White Field, Bangalore, 560066, India; Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, 690525, India
| | - Gourav Dey
- Institute of Bioinformatics, International Technology Park, White Field, Bangalore, 560066, India
| | - Firdous Ahmad Bhat
- Institute of Bioinformatics, International Technology Park, White Field, Bangalore, 560066, India
| | - Oishi Chatterjee
- Institute of Bioinformatics, International Technology Park, White Field, Bangalore, 560066, India; Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, 690525, India
| | - Sayali Chandrashekhar Deolankar
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, 575018, India
| | - Mariamma Philip
- Department of Biostatistics, NIMHANS, No. 2900, Hosur Road, Bangalore, 560029, India
| | - T S Keshava Prasad
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, 575018, India.
| | - M M Srinivas Bharath
- Department of Clinical Psychopharmacology and Neurotoxicology, NIMHANS, No. 2900, Hosur Road, Bangalore, 560029, India.
| | - Anita Mahadevan
- Department of Neuropathology, National Institute of Mental Health and Neurosciences (NIMHANS), No. 2900, Hosur Road, Bangalore, 560029, India.
| |
Collapse
|
14
|
Niotis K, Akiyoshi K, Carlton C, Isaacson R. Dementia Prevention in Clinical Practice. Semin Neurol 2022; 42:525-548. [PMID: 36442814 DOI: 10.1055/s-0042-1759580] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Over 55 million people globally are living with dementia and, by 2050, this number is projected to increase to 131 million. This poses immeasurable challenges for patients and their families and a significant threat to domestic and global economies. Given this public health crisis and disappointing results from disease-modifying trials, there has been a recent shift in focus toward primary and secondary prevention strategies. Approximately 40% of Alzheimer's disease (AD) cases, which is the most common form of dementia, may be prevented or at least delayed. Success of risk reduction studies through addressing modifiable risk factors, in addition to the failure of most drug trials, lends support for personalized multidomain interventions rather than a "one-size-fits-all" approach. Evolving evidence supports early intervention in at-risk patients using individualized interventions directed at modifiable risk factors. Comprehensive risk stratification can be informed by emerging principals of precision medicine, and include expanded clinical and family history, anthropometric measurements, blood biomarkers, neurocognitive evaluation, and genetic information. Risk stratification is key in differentiating subtypes of dementia and identifies targetable areas for intervention. This article reviews a clinical approach toward dementia risk stratification and evidence-based prevention strategies, with a primary focus on AD.
Collapse
Affiliation(s)
- Kellyann Niotis
- Department of Neurology, Weill Cornell Medicine and New York - Presbyterian, New York, New York
| | - Kiarra Akiyoshi
- Department of Neurology, Weill Cornell Medicine and New York - Presbyterian, New York, New York
| | - Caroline Carlton
- Department of Neurology, Weill Cornell Medicine and New York - Presbyterian, New York, New York
| | - Richard Isaacson
- Department of Neurology, Weill Cornell Medicine and New York - Presbyterian, New York, New York.,Department of Neurology, Florida Atlantic University, Charles E. Schmidt College of Medicine, Boca Raton, Florida
| |
Collapse
|
15
|
Lipolysis-Stimulated Lipoprotein Receptor Acts as Sensor to Regulate ApoE Release in Astrocytes. Int J Mol Sci 2022; 23:ijms23158630. [PMID: 35955777 PMCID: PMC9368974 DOI: 10.3390/ijms23158630] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/28/2022] [Accepted: 07/30/2022] [Indexed: 11/17/2022] Open
Abstract
Astroglia play an important role, providing de novo synthesized cholesterol to neurons in the form of ApoE-lipidated particles; disruption of this process can increase the risk of Alzheimer’s disease. We recently reported that glia-specific suppression of the lipolysis-stimulated lipoprotein receptor (LSR) gene leads to Alzheimer’s disease-like memory deficits. Since LSR is an Apo-E lipoprotein receptor, our objective of this study was to determine the effect of LSR expression modulation on cholesterol and ApoE output in mouse astrocytes expressing human ApoE3. qPCR analysis showed that siRNA-mediated lsr knockdown significantly increased expression of the genes involved in cholesterol synthesis, secretion, and metabolism. Analysis of media and lipoprotein fractions showed increased cholesterol and lipidated ApoE output in HDL-like particles. Further, lsr expression could be upregulated when astrocytes were incubated 5 days in media containing high levels (two-fold) of lipoprotein, or after 8 h treatment with 1 µM LXR agonist T0901317 in lipoprotein-deficient media. In both conditions of increased lsr expression, the ApoE output was repressed or unchanged despite increased abca1 mRNA levels and cholesterol production. We conclude that LSR acts as a sensor of lipoprotein content in the medium and repressor of ApoE release, while ABCA1 drives cholesterol efflux, thereby potentially affecting cholesterol load, ApoE lipidation, and limiting cholesterol trafficking towards the neuron.
Collapse
|
16
|
Ding Y, Wang L, Sun J, Shi Y, Li G, Luan X, Zheng G, Zhang G. Remnant Cholesterol and Dyslipidemia Are Risk Factors for Guillain–Barré Syndrome and Severe Guillain–Barré Syndrome by Promoting Monocyte Activation. Front Immunol 2022; 13:946825. [PMID: 35911688 PMCID: PMC9326451 DOI: 10.3389/fimmu.2022.946825] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 06/21/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundGuillain–Barré syndrome (GBS) is the most common severe acute paralytic neuropathy, with a mortality rate of 5% and permanent sequelae rate of 10%. Currently, the cause of GBS remains unclear. Therefore, we sought to determine potential predictors for GBS and its severity.MethodsA case–control study was performed at Tiantan Hospital in Beijing from January 2017 to December 2021. Laboratory and clinical characteristics were assessed in recruited GBS patients and healthy control individuals (matched by sex and age). The potential risk factors for GBS and severe GBS were assessed using a logistic regression analysis. The mRNA levels of toll-like receptor 4 (TLR4), toll-like receptor 2 (TLR2) and nuclear factor κB (NF-κB) in GBS patients and control PBMCs were detected by fluorescence quantitative PCR. THP-1 cells were costimulated with LPS and free cholesterol to demonstrate the effect of free cholesterol on monocyte activation.ResultsA total of 147 GBS patients and 153 healthy individuals were included in the study. Logistic regression analyses showed that preceding infection, alcohol consumption, remnant cholesterol, homocysteine and the dyslipidemia index were correlated with a higher risk of GBS. In contrast, increased HDL cholesterol was correlated with a lower risk of GBS. Moreover, remnant cholesterol and the dyslipidemia index were significantly correlated with severe GBS. The mRNA levels of TLR4, TLR2 and NF-κB in the PBMCs of GBS patients were significantly higher than those of healthy individuals. LPS activated THP-1 cells, and free cholesterol treatment increased the expression of TLR4, TLR2, NF-κB and IL-1β mRNA in LPS-activated THP-1 cells.ConclusionDyslipidemia was correlated with the risk of GBS and severe GBS. Remnant cholesterol may promote the activation of monocytes in GBS patients. It may be valuable to control lipid levels in the prevention of GBS and severe GBS.
Collapse
Affiliation(s)
- Yaowei Ding
- Department of Clinical Diagnosis, Laboratory of Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Lijuan Wang
- Department of Clinical Diagnosis, Laboratory of Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jialu Sun
- Department of Clinical Diagnosis, Laboratory of Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yijun Shi
- Department of Clinical Diagnosis, Laboratory of Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Guoge Li
- Department of Clinical Diagnosis, Laboratory of Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xin Luan
- Department of Clinical Diagnosis, Laboratory of Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Guanghui Zheng
- Department of Clinical Diagnosis, Laboratory of Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- National Medical Products Administration (NMPA) Key Laboratory for Quality Control of In Vitro Diagnostics, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Beijing Engineering Research Center of Immunological Reagents Clinical Research, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Guojun Zhang
- Department of Clinical Diagnosis, Laboratory of Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- National Medical Products Administration (NMPA) Key Laboratory for Quality Control of In Vitro Diagnostics, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Beijing Engineering Research Center of Immunological Reagents Clinical Research, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- *Correspondence: Guojun Zhang,
| |
Collapse
|
17
|
Tong JH, Gong SQ, Zhang YS, Dong JR, Zhong X, Wei MJ, Liu MY. Association of Circulating Apolipoprotein AI Levels in Patients With Alzheimer's Disease: A Systematic Review and Meta-Analysis. Front Aging Neurosci 2022; 14:899175. [PMID: 35663584 PMCID: PMC9157647 DOI: 10.3389/fnagi.2022.899175] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 04/19/2022] [Indexed: 11/26/2022] Open
Abstract
With the development of medicine, our research on Alzheimer's disease (AD) has been further deepened, but the mechanism of its occurrence and development has not been fully revealed, and there is currently no effective treatment method. Several studies have shown that apolipoprotein AI (ApoA-I) can affect the occurrence and development of Alzheimer's disease by binding to amyloid β (Aβ). However, the association between circulating levels of ApoA-I and AD remains controversial. We conducted a meta-analysis of 18 studies published between 1992 and 2017 to determine whether the ApoA-I levels in the blood and cerebrospinal fluid (CSF) are abnormal in AD. Literatures were searched in PubMed, EMBASE and Web of Science databases without language limitations. A pooled subject sample including 1,077 AD patients and 1,271 healthy controls (HCs) was available to assess circulating ApoA-I levels; 747 AD patients and 680 HCs were included for ApoA-I levels in serum; 246 AD patients and 456 HCs were included for ApoA-I levels in plasma; 201 AD patients and 447 HCs were included for ApoA-I levels in CSF. It was found that serum and plasma levels of ApoA-I were significantly reduced in AD patients compared with HCs {[standardized mean difference (SMD) = −1.16; 95% confidence interval (CI) (−1.72, −0.59); P = 0.000] and [SMD = −1.13; 95% CI (−2.05, −0.21); P = 0.016]}. Patients with AD showed a tendency toward higher CSF ApoA-I levels compared with HCs, although this difference was non-significant [SMD = 0.20; 95% CI (−0.16, 0.56); P = 0.273]. In addition, when we analyzed the ApoA-I levels of serum and plasma together, the circulating ApoA-I levels in AD patients was significantly lower [SMD = −1.15; 95% CI (−1.63, −0.66); P = 0.000]. These results indicate that ApoA-I deficiency may be a risk factor of AD, and ApoA-I has the potential to serve as a biomarker for AD and provide experimental evidence for diagnosis of AD. Systematic Review Registration: PROSPERO, identifier: 325961.
Collapse
|
18
|
Uchida K. Waste Clearance in the Brain and Neuroinflammation: A Novel Perspective on Biomarker and Drug Target Discovery in Alzheimer's Disease. Cells 2022; 11:cells11050919. [PMID: 35269541 PMCID: PMC8909773 DOI: 10.3390/cells11050919] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/26/2022] [Accepted: 03/04/2022] [Indexed: 02/06/2023] Open
Abstract
Alzheimer’s disease (AD) is a multifactorial disease with a heterogeneous etiology. The pathology of Alzheimer’s disease is characterized by amyloid-beta and hyperphosphorylated tau, which are necessary for disease progression. Many clinical trials on disease-modifying drugs for AD have failed to indicate their clinical benefits. Recent advances in fundamental research have indicated that neuroinflammation plays an important pathological role in AD. Damage- and pathogen-associated molecular patterns in the brain induce neuroinflammation and inflammasome activation, causing caspase-1-dependent glial and neuronal cell death. These waste products in the brain are eliminated by the glymphatic system via perivascular spaces, the blood-brain barrier, and the blood–cerebrospinal fluid barrier. Age-related vascular dysfunction is associated with an impairment of clearance and barrier functions, leading to neuroinflammation. The proteins involved in waste clearance in the brain and peripheral circulation may be potential biomarkers and drug targets in the early stages of cognitive impairment. This short review focuses on waste clearance dysfunction in AD pathobiology and discusses the improvement of waste clearance as an early intervention in prodromal AD and preclinical stages of dementia.
Collapse
Affiliation(s)
- Kazuhiko Uchida
- Faculty of Medicine, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba 305-8575, Ibaraki, Japan; ; Tel.: +81-29-853-3210; Fax: +81-50-3730-7456
- Institute for Biomedical Research, MCBI, 4-9-29 Matsushiro, Tsukuba 305-0035, Ibaraki, Japan
| |
Collapse
|
19
|
Emerging role of HDL in brain cholesterol metabolism and neurodegenerative disorders. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159123. [PMID: 35151900 DOI: 10.1016/j.bbalip.2022.159123] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 02/01/2022] [Accepted: 02/03/2022] [Indexed: 01/07/2023]
Abstract
High-density lipoproteins (HDLs play a key role in cholesterol homeostasis maintenance in the central nervous system (CNS), by carrying newly synthesized cholesterol from astrocytes to neurons, to support their lipid-related physiological functions. As occurs for plasma HDLs, brain lipoproteins are assembled through the activity of membrane cholesterol transporters, undergo remodeling mediated by specific enzymes and transport proteins, and finally deliver cholesterol to neurons by a receptor-mediated internalization process. A growing number of evidences indicates a strong association between alterations of CNS cholesterol homeostasis and neurodegenerative disorders, in particular Alzheimer's disease (AD), and a possible role in this relationship may be played by defects in brain HDL metabolism. In the present review, we summarize and critically examine the current state of knowledge on major modifications of HDL and HDL-mediated brain cholesterol transport in AD, by taking into consideration the individual steps of this process. We also describe potential and encouraging HDL-based therapies that could represent new therapeutic strategies for AD treatment. Finally, we revise the main plasma and brain HDL modifications in other neurodegenerative disorders including Parkinson's disease (PD), Huntington's disease (HD), and frontotemporal dementia (FTD).
Collapse
|
20
|
Kundu P, Zimmerman B, Quinn JF, Kaye J, Mattek N, Westaway SK, Raber J. Serum Levels of α-Klotho Are Correlated with Cerebrospinal Fluid Levels and Predict Measures of Cognitive Function. J Alzheimers Dis 2022; 86:1471-1481. [PMID: 35213382 PMCID: PMC9108571 DOI: 10.3233/jad-215719] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2022] [Indexed: 02/05/2023]
Abstract
BACKGROUND α-klotho might play a role in neurodegenerative diseases. OBJECTIVE To determine levels of α-klotho and apoE in serum and cerebrospinal fluid (CSF) samples and their relationship with the Mini-Mental State Examination (MMSE) and Clinical Dementia Rating (CDR). METHODS All subjects were between age 39 to 83+ (n = 94). CDR and MMSE were administered to all participants. CSF was collected in the early afternoon by lumbar puncture. RESULTS Serum and CSF levels of α-klotho are positively correlated and both predict scores on the MMSE and CDR, regardless of sex or apoE4 status. CONCLUSION Our results demonstrate that α-klotho may be an important biomarker of cognitive health and neurodegeneration, and that relatively non-invasive sampling of α-klotho from serum is likely highly reflective of CSF levels.
Collapse
Affiliation(s)
- Payel Kundu
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, USA
| | - Benjamin Zimmerman
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, USA
- Advanced Imaging Research Center, Oregon Health and Science University, Portland, OR, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Joseph F. Quinn
- Department of Neurology, Oregon Health and Science University, Portland, OR, USA
- Department of Neurology, Veterans Affairs Medical Center, Portland, OR, USA
| | - Jeffrey Kaye
- Department of Neurology, Oregon Health and Science University, Portland, OR, USA
| | - Nora Mattek
- Department of Neurology, Oregon Health and Science University, Portland, OR, USA
| | - Shawn K. Westaway
- Department of Neurology, Oregon Health and Science University, Portland, OR, USA
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, USA
- Department of Neurology, Oregon Health and Science University, Portland, OR, USA
- Departments of Psychiatry and Radiation Medicine, Division of Neuroscience, ONPRC, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
21
|
Sierri G, Dal Magro R, Vergani B, Leone BE, Formicola B, Taiarol L, Fagioli S, Kravicz M, Tremolizzo L, Calabresi L, Re F. Reduced Levels of ABCA1 Transporter Are Responsible for the Cholesterol Efflux Impairment in β-Amyloid-Induced Reactive Astrocytes: Potential Rescue from Biomimetic HDLs. Int J Mol Sci 2021; 23:ijms23010102. [PMID: 35008528 PMCID: PMC8745016 DOI: 10.3390/ijms23010102] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/16/2021] [Accepted: 12/20/2021] [Indexed: 12/02/2022] Open
Abstract
The cerebral synthesis of cholesterol is mainly handled by astrocytes, which are also responsible for apoproteins’ synthesis and lipoproteins’ assembly required for the cholesterol transport in the brain parenchyma. In Alzheimer disease (AD), these processes are impaired, likely because of the astrogliosis, a process characterized by morphological and functional changes in astrocytes. Several ATP-binding cassette transporters expressed by brain cells are involved in the formation of nascent discoidal lipoproteins, but the effect of beta-amyloid (Aβ) assemblies on this process is not fully understood. In this study, we investigated how of Aβ1-42-induced astrogliosis affects the metabolism of cholesterol in vitro. We detected an impairment in the cholesterol efflux of reactive astrocytes attributable to reduced levels of ABCA1 transporters that could explain the decreased lipoproteins’ levels detected in AD patients. To approach this issue, we designed biomimetic HDLs and evaluated their performance as cholesterol acceptors. The results demonstrated the ability of apoA-I nanodiscs to cross the blood–brain barrier in vitro and to promote the cholesterol efflux from astrocytes, making them suitable as a potential supportive treatment for AD to compensate the depletion of cerebral HDLs.
Collapse
Affiliation(s)
- Giulia Sierri
- BioNanoMedicine Center NANOMIB, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (G.S.); (R.D.M.); (B.F.); (L.T.); (S.F.); (M.K.)
| | - Roberta Dal Magro
- BioNanoMedicine Center NANOMIB, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (G.S.); (R.D.M.); (B.F.); (L.T.); (S.F.); (M.K.)
| | - Barbara Vergani
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (B.V.); (B.E.L.); (L.T.)
| | - Biagio Eugenio Leone
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (B.V.); (B.E.L.); (L.T.)
| | - Beatrice Formicola
- BioNanoMedicine Center NANOMIB, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (G.S.); (R.D.M.); (B.F.); (L.T.); (S.F.); (M.K.)
| | - Lorenzo Taiarol
- BioNanoMedicine Center NANOMIB, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (G.S.); (R.D.M.); (B.F.); (L.T.); (S.F.); (M.K.)
| | - Stefano Fagioli
- BioNanoMedicine Center NANOMIB, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (G.S.); (R.D.M.); (B.F.); (L.T.); (S.F.); (M.K.)
| | - Marcelo Kravicz
- BioNanoMedicine Center NANOMIB, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (G.S.); (R.D.M.); (B.F.); (L.T.); (S.F.); (M.K.)
| | - Lucio Tremolizzo
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (B.V.); (B.E.L.); (L.T.)
| | - Laura Calabresi
- Department of Pharmacological and Biomolecular Science, Centro Grossi Paoletti, University of Milan, 20133 Milan, Italy;
| | - Francesca Re
- BioNanoMedicine Center NANOMIB, School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (G.S.); (R.D.M.); (B.F.); (L.T.); (S.F.); (M.K.)
- Correspondence:
| |
Collapse
|
22
|
Endres K. Apolipoprotein A1, the neglected relative of Apolipoprotein E and its potential role in Alzheimer's disease. Neural Regen Res 2021; 16:2141-2148. [PMID: 33818485 PMCID: PMC8354123 DOI: 10.4103/1673-5374.310669] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/22/2020] [Accepted: 02/02/2021] [Indexed: 01/23/2023] Open
Abstract
Lipoproteins are multi-molecule assemblies with the primary function of transportation and processing of lipophilic substances within aqueous bodily fluids (blood, cerebrospinal fluid). Nevertheless, they also exert other physiological functions such as immune regulation. In particular, neurons are both sensitive to uncontrolled responses of the immune system and highly dependent on a controlled and sufficient supply of lipids. For this reason, the role of certain lipoproteins and their protein-component (apolipoproteins, Apo's) in neurological diseases is perceivable. ApoE, for example, is well-accepted as one of the major risk factors for sporadic Alzheimer's disease with a protective allele variant (ε2) and a risk-causing allele variant (ε4). ApoA1, the major protein component of high-density lipoproteins, is responsible for transportation of excess cholesterol from peripheral tissues to the liver. The protein is synthesized in the liver and intestine but also can enter the brain via the choroid plexus and thereby might have an impact on brain lipid homeostasis. This review focuses on the role of ApoA1 in Alzheimer's disease and discusses whether its role within this neurodegenerative disorder is specific or represents a general neuroprotective mechanism.
Collapse
Affiliation(s)
- Kristina Endres
- Department of Psychiatry and Psychotherapy, University Medical Center of the Johannes Gutenberg-University Mainz, Untere Zahlbacher Str. 8, 55131 Mainz, Germany
| |
Collapse
|
23
|
Esteban de Antonio E, Pérez-Cordón A, Gil S, Orellana A, Cano A, Alegret M, Espinosa A, Alarcón-Martín E, Valero S, Martínez J, de Rojas I, Sotolongo-Grau Ó, Martín E, Vivas A, Gomez-Chiari M, Tejero MÁ, Bernuz M, Tárraga L, Ruiz A, Marquié M, Boada M. BIOFACE: A Prospective Study of Risk Factors, Cognition, and Biomarkers in a Cohort of Individuals with Early-Onset Mild Cognitive Impairment. Study Rationale and Research Protocols. J Alzheimers Dis 2021; 83:1233-1249. [PMID: 34420953 PMCID: PMC8543256 DOI: 10.3233/jad-210254] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Background: Mild cognitive impairment (MCI) due to Alzheimer’s disease (AD) diagnosis is based on cerebrospinal fluid (CSF) or neuroimaging biomarkers. Currently, non-invasive and inexpensive blood-based biomarkers are being investigated, such as neuronal-derived plasma exosomes (NPEs). Neuroinflammation and early vascular changes have been described in AD pathogenesis and can be traced in plasma and NPEs. However, they have not been studied in early onset MCI (EOMCI). Objective: To describe the rationale, design, and baseline characteristics of the participants from the BIOFACE cohort, a two-year observational study on EOMCI conducted at Fundació ACE. The study goal is to characterize the different phenotypes from a clinical, neuropsychological, and biomarker point of view and to investigate the CSF and plasma proteomics as well as the role of NPEs as early biomarkers of AD. Methods: Participants underwent extended neurological and neuropsychological batteries, multimodal biomarkers including brain MRI, blood, saliva, CSF, anthropometric, and neuro-ophthalmological examinations. Results: Ninety-seven patients with EOMCI were recruited. 59.8%were women. Mean age at symptom onset was 57 years; mean MMSE was 28. First degree and presenile family history of dementia was present in 60.8%and 15.5%, respectively. Depressive and anxiety disorders along with vascular risk factors were the most frequent comorbidities. 29%of participants were APOE ɛ4 carriers, and 67%showed a CSF normal ATN profile. Conclusion: BIOFACE is a two-year study of clinical, cognition, and biomarkers that will shed light on the physiopathology and the potential utility of plasma and NPEs as non-invasive early diagnostic and prognostic biomarkers in people younger than 65 years.
Collapse
Affiliation(s)
- Ester Esteban de Antonio
- Research Center and Memory Clinic, Fundació ACE, Institut Catalá de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Alba Pérez-Cordón
- Research Center and Memory Clinic, Fundació ACE, Institut Catalá de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Silvia Gil
- Research Center and Memory Clinic, Fundació ACE, Institut Catalá de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Adelina Orellana
- Research Center and Memory Clinic, Fundació ACE, Institut Catalá de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Amanda Cano
- Research Center and Memory Clinic, Fundació ACE, Institut Catalá de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Montserrat Alegret
- Research Center and Memory Clinic, Fundació ACE, Institut Catalá de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Ana Espinosa
- Research Center and Memory Clinic, Fundació ACE, Institut Catalá de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Emilio Alarcón-Martín
- Research Center and Memory Clinic, Fundació ACE, Institut Catalá de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Sergi Valero
- Research Center and Memory Clinic, Fundació ACE, Institut Catalá de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Joan Martínez
- Research Center and Memory Clinic, Fundació ACE, Institut Catalá de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Itziar de Rojas
- Research Center and Memory Clinic, Fundació ACE, Institut Catalá de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Óscar Sotolongo-Grau
- Research Center and Memory Clinic, Fundació ACE, Institut Catalá de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Elvira Martín
- Research Center and Memory Clinic, Fundació ACE, Institut Catalá de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain
| | - Assumpta Vivas
- Departament de Diagnòstic per la Imatge, Clínica Corachan, Barcelona, Spain
| | - Marta Gomez-Chiari
- Departament de Diagnòstic per la Imatge, Clínica Corachan, Barcelona, Spain
| | | | - Mireia Bernuz
- Grup de Sensors i Biosensors, Departament de Química, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Lluis Tárraga
- Research Center and Memory Clinic, Fundació ACE, Institut Catalá de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Agustín Ruiz
- Research Center and Memory Clinic, Fundació ACE, Institut Catalá de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Marquié
- Research Center and Memory Clinic, Fundació ACE, Institut Catalá de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Mercè Boada
- Research Center and Memory Clinic, Fundació ACE, Institut Catalá de Neurociències Aplicades, Universitat Internacional de Catalunya, Barcelona, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | | |
Collapse
|
24
|
Ferguson AC, Tank R, Lyall LM, Ward J, Celis-Morales C, Strawbridge R, Ho F, Whelan CD, Gill J, Welsh P, Anderson JJ, Mark PB, Mackay DF, Smith DJ, Pell JP, Cavanagh J, Sattar N, Lyall DM. Alzheimer's Disease Susceptibility Gene Apolipoprotein E (APOE) and Blood Biomarkers in UK Biobank (N = 395,769). J Alzheimers Dis 2021; 76:1541-1551. [PMID: 32651323 DOI: 10.3233/jad-200338] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurodegenerative condition where the underlying etiology is still unclear. Investigating the potential influence of apolipoprotein E (APOE), a major genetic risk factor, on common blood biomarkers could provide a greater understanding of the mechanisms of AD and dementia risk. OBJECTIVE Our objective was to conduct the largest (to date) single-protocol investigation of blood biomarkers in the context of APOE genotype, in UK Biobank. METHODS After quality control and exclusions, data on 395,769 participants of White European ancestry were available for analysis. Linear regressions were used to test potential associations between APOE genotypes and biomarkers. RESULTS Several biomarkers significantly associated with APOEɛ4 'risk' and ɛ2 'protective' genotypes (versus neutral ɛ3/ɛ3). Most associations supported previous data: for example, ɛ4 genotype was associated with elevated low-density lipoprotein cholesterol (LDL) (standardized beta [b] = 0.150 standard deviations [SDs] per allele, p < 0.001) and ɛ2 with lower LDL (b = -0.456 SDs, p < 0.001). There were however instances of associations found in unexpected directions: e.g., ɛ4 and increased insulin-like growth factor (IGF-1) (b = 0.017, p < 0.001) where lower levels have been previously suggested as an AD risk factor. CONCLUSION These findings highlight biomarker differences in non-demented people at genetic risk for dementia. The evidence herein supports previous hypotheses of involvement from cardiometabolic and neuroinflammatory pathways.
Collapse
Affiliation(s)
- Amy C Ferguson
- Institute of Health and Wellbeing, University of Glasgow, Scotland, UK
| | - Rachana Tank
- Institute of Health and Wellbeing, University of Glasgow, Scotland, UK
| | - Laura M Lyall
- Institute of Health and Wellbeing, University of Glasgow, Scotland, UK
| | - Joey Ward
- Institute of Health and Wellbeing, University of Glasgow, Scotland, UK
| | - Carlos Celis-Morales
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland, UK.,GEEAFyS, Universidad Católica del Maule, Talca, Chile
| | - Rona Strawbridge
- Institute of Health and Wellbeing, University of Glasgow, Scotland, UK.,Health Data Research UK.,Department of Medicine Solna, Cardiovascular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Frederick Ho
- Institute of Health and Wellbeing, University of Glasgow, Scotland, UK
| | | | - Jason Gill
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland, UK
| | - Paul Welsh
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland, UK
| | - Jana J Anderson
- Institute of Health and Wellbeing, University of Glasgow, Scotland, UK
| | - Patrick B Mark
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, Scotland, UK
| | - Daniel F Mackay
- Institute of Health and Wellbeing, University of Glasgow, Scotland, UK
| | - Daniel J Smith
- Institute of Health and Wellbeing, University of Glasgow, Scotland, UK
| | - Jill P Pell
- Institute of Health and Wellbeing, University of Glasgow, Scotland, UK
| | - Jonathan Cavanagh
- Institute of Health and Wellbeing, University of Glasgow, Scotland, UK
| | - Naveed Sattar
- Institute of Health and Wellbeing, University of Glasgow, Scotland, UK
| | - Donald M Lyall
- Institute of Health and Wellbeing, University of Glasgow, Scotland, UK
| |
Collapse
|
25
|
Zuin M, Cervellati C, Trentini A, Passaro A, Rosta V, Zimetti F, Zuliani G. Association between Serum Concentrations of Apolipoprotein A-I (ApoA-I) and Alzheimer's Disease: Systematic Review and Meta-Analysis. Diagnostics (Basel) 2021; 11:984. [PMID: 34071695 PMCID: PMC8229134 DOI: 10.3390/diagnostics11060984] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/26/2021] [Accepted: 05/27/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND A wealth of experimental and epidemiological evidence suggest that Apolipoprotein A-I (ApoA-I), the main protein constituent of high-density lipoprotein (HDL), may protect against Alzheimer disease (AD). To investigate this potential role, we conducted a meta-analysis of the published studies on the relationship between serum ApoA-I and AD occurrence. METHODS We screened MEDLINE, EMBASE, Web of Science, and Scopus, for cross-sectional studies published from inception to 1 March 2021, comparing the ApoA-I serum levels between patients with AD and cognitively normal controls. RESULTS From an initial screening of 245 articles, 5 studies, including 397 AD patients (mean age 75.0 years, 234 females) and 367 controls (mean age 69.2 years, 182 females), met the inclusion criteria. Compared to healthy controls, AD subjects had a lower ApoA-I serum level. The pooled weighted mean difference from a random-effects model was -0.31 g/L (p < 0.0001) (95% Confidence Interval: [-0.62-0.01], with high heterogeneity (I2 = 100%). The Egger's test confirmed an absence of publication bias (t = 0.62, p = 0.576). CONCLUSIONS Our study showed that AD patients present lower serum levels of ApoA-I compared to cognitively normal individuals. Further studies on large population samples are required to support this finding.
Collapse
Affiliation(s)
- Marco Zuin
- Department of Translational Medicine and for Romagna, University of Ferrara, 44121 Ferrara, Italy
| | - Carlo Cervellati
- Department of Translational Medicine and for Romagna, University of Ferrara, 44121 Ferrara, Italy
| | - Alessandro Trentini
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Angelina Passaro
- Department of Translational Medicine and for Romagna, University of Ferrara, 44121 Ferrara, Italy
| | - Valentina Rosta
- Department of Translational Medicine and for Romagna, University of Ferrara, 44121 Ferrara, Italy
| | - Francesca Zimetti
- Department of Food and Drug, University of Parma, 43121 Parma, Italy
| | - Giovanni Zuliani
- Department of Translational Medicine and for Romagna, University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
26
|
Azizidoost SH, Babaahmadi-Rezaei H, Nazeri Z, Cheraghzadeh M, Kheirollah A. Impact of Methyl-β-Cyclodextrin and Apolipoprotein A-I on The Expression of ATP-Binding Cassette Transporter A1 and Cholesterol Depletion in C57BL/6 Mice Astrocytes. CELL JOURNAL 2021; 23:93-98. [PMID: 33650825 PMCID: PMC7944131 DOI: 10.22074/cellj.2021.7061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 11/25/2019] [Indexed: 11/04/2022]
Abstract
Objective Dysregulation of cholesterol metabolism in the brain is responsible for many lipid storage disorders, including
Niemann-Pick disease type C (NPC). Here, we have investigated whether cyclodextrin (CD) and apolipoprotein A-I
(apoA-I) induce the same signal to inhibit cell cholesterol accumulation by focusing on the main proteins involved in
cholesterol homeostasis in response to CD and apoA-I treatment.
Materials and Methods In this experimental study, astrocytes were treated with apoA-I or CD and then lysed in RIPA
buffer. We used Western blot to detect protein levels of 3-hydroxy-3-methyl-glutaryl coenzyme A reductase (HMGCR)
and ATP-binding cassette transporter A1 (ABCA1). Cell cholesterol content and cholesterol release in the medium were
also measured.
Results ApoA-I induced a significant increase in ABCA1 and a mild increase in HMGCR protein level, whereas
CD caused a significant increase in HMGCR with a significant decrease in ABCA1. Both apoA-I and CD increased
cholesterol release in the medium. A mild, but not significant increase, in cell cholesterol content was seen by apoA-I;
however, a significant increase in cell cholesterol was detected when the astrocytes were treated with CD.
Conclusion CD, like apoA-I, depletes cellular cholesterol. This depletion occurs in a different way from apoA-I that
is through cholesterol efflux. Depletion of cell cholesterol with CDs led to reduced protein levels of ABCA1 along with
increased HMGCR and accumulation of cell cholesterol. This suggested that CDs, unlike apoA-I, could impair the
balance between cholesterol synthesis and release, and interfere with cellular function that depends on ABCA1.
Collapse
Affiliation(s)
- S Hirin Azizidoost
- Cellular and Molecular Research Center, Department of Biochemistry, Medical School, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hossein Babaahmadi-Rezaei
- Cellular and Molecular Research Center, Department of Biochemistry, Medical School, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Zahra Nazeri
- Cellular and Molecular Research Center, Department of Biochemistry, Medical School, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Maryam Cheraghzadeh
- Cellular and Molecular Research Center, Department of Biochemistry, Medical School, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Alireza Kheirollah
- Cellular and Molecular Research Center, Department of Biochemistry, Medical School, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
27
|
Pillai JA, Bena J, Bekris LM, Foldvary-Schaefer N, Heinzinger C, Rao S, Rao SM, Leverenz JB, Mehra R. Unique Sleep and Circadian Rhythm Dysfunction Neuroinflammatory and Immune Profiles in Alzheimer's Disease with Mild Cognitive Impairment. J Alzheimers Dis 2021; 81:487-492. [PMID: 33814445 PMCID: PMC8179975 DOI: 10.3233/jad-201573] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Sleep dysfunction has been identified in the pathophysiology of Alzheimer's disease (AD); however, the role and mechanism of circadian rhythm dysfunction is less well understood. In a well-characterized cohort of patients with AD at the mild cognitive impairment stage (MCI-AD), we identify that circadian rhythm irregularities were accompanied by altered humoral immune responses detected in both the cerebrospinal fluid and plasma as well as alterations of cerebrospinal fluid biomarkers of neurodegeneration. On the other hand, sleep disruption was more so associated with abnormalities in circulating markers of immunity and inflammation and decrements in cognition.
Collapse
Affiliation(s)
- Jagan A. Pillai
- Lou Ruvo Center for Brain Health, Cleveland Clinic, Cleveland, OH, USA
- Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Neurology, Cleveland Clinic, Cleveland, OH, USA
| | - James Bena
- Quantitative Health Sciences, Cleveland Clinic, Cleveland, OH, USA
| | - Lynn M. Bekris
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
| | | | - Catherine Heinzinger
- Sleep Disorders Center, Neurologic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Sujata Rao
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Stephen M. Rao
- Lou Ruvo Center for Brain Health, Cleveland Clinic, Cleveland, OH, USA
- Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - James B. Leverenz
- Lou Ruvo Center for Brain Health, Cleveland Clinic, Cleveland, OH, USA
- Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Neurology, Cleveland Clinic, Cleveland, OH, USA
| | - Reena Mehra
- Sleep Disorders Center, Neurologic Institute, Cleveland Clinic, Cleveland, OH, USA
- Respiratory Institute, Cleveland Clinic, Cleveland, OH, USA
- Heart and Vascular Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
28
|
Marsillach J, Adorni MP, Zimetti F, Papotti B, Zuliani G, Cervellati C. HDL Proteome and Alzheimer's Disease: Evidence of a Link. Antioxidants (Basel) 2020; 9:E1224. [PMID: 33287338 PMCID: PMC7761753 DOI: 10.3390/antiox9121224] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/25/2020] [Accepted: 11/30/2020] [Indexed: 12/16/2022] Open
Abstract
Several lines of epidemiological evidence link increased levels of high-density lipoprotein-cholesterol (HDL-C) with lower risk of Alzheimer's disease (AD). This observed relationship might reflect the beneficial effects of HDL on the cardiovascular system, likely due to the implication of vascular dysregulation in AD development. The atheroprotective properties of this lipoprotein are mostly due to its proteome. In particular, apolipoprotein (Apo) A-I, E, and J and the antioxidant accessory protein paraoxonase 1 (PON1), are the main determinants of the biological function of HDL. Intriguingly, these HDL constituent proteins are also present in the brain, either from in situ expression, or derived from the periphery. Growing preclinical evidence suggests that these HDL proteins may prevent the aberrant changes in the brain that characterize AD pathogenesis. In the present review, we summarize and critically examine the current state of knowledge on the role of these atheroprotective HDL-associated proteins in AD pathogenesis and physiopathology.
Collapse
Affiliation(s)
- Judit Marsillach
- Department of Environmental & Occupational Health Sciences, University of Washington, Seattle, WA 98195, USA;
| | - Maria Pia Adorni
- Unit of Neurosciences, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy;
| | - Francesca Zimetti
- Department of Food and Drug, University of Parma, 43124 Parma, Italy;
| | - Bianca Papotti
- Department of Food and Drug, University of Parma, 43124 Parma, Italy;
| | - Giovanni Zuliani
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (G.Z.); (C.C.)
| | - Carlo Cervellati
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (G.Z.); (C.C.)
| |
Collapse
|
29
|
Bradley D. Clusterin as a Potential Biomarker of Obesity-Related Alzheimer's Disease Risk. Biomark Insights 2020; 15:1177271920964108. [PMID: 33110346 PMCID: PMC7555556 DOI: 10.1177/1177271920964108] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 09/14/2020] [Indexed: 02/03/2023] Open
Abstract
Over 35% of the adult US population is obese. In turn, excess adiposity increases the risk of multiple complications including type 2 diabetes (T2D), insulin resistance, and cardiovascular disease; yet, obesity also independently heightens risk of Alzheimer's Disease (AD), even after adjusting for other important confounding risk factors including blood pressure, sociodemographics, cholesterol levels, smoking status, and Apolipoprotein E (ApoE) genotype. Among patients over the age of 65 with dementia, 37% have coexisting diabetes, and an estimated 7.3% of cases of AD are directly attributable to midlife obesity. Clusterin, also known as apolipoprotein J (ApoJ), is a multifunctional glycoprotein that acts as a molecular chaperone, assisting folding of secreted proteins. Clusterin has been implicated in several physiological and pathological states, including AD, metabolic disease, and cardiovascular disease. Despite long-standing interest in elucidating clusterin's relationship with amyloid beta (Aβ) aggregation/clearance and toxicity, significant knowledge gaps still exist. Altered clusterin expression and protein levels have been linked with cognitive and memory function, disrupted central nervous system lipid flux, as well as pathogenic brain structure; and its role in cardiometabolic disease suggests that it may be a link between insulin resistance, dyslipidemia, and AD. Here, we briefly highlight clusterin's relevance to AD by presenting existing evidence linking clusterin to AD and cardiometabolic disease, and discussing its potential utility as a biomarker for AD in the presence of obesity-related metabolic disease.
Collapse
Affiliation(s)
- David Bradley
- Diabetes and Metabolism Research Center, Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
30
|
Loupy KM, Lee T, Zambrano CA, Elsayed AI, D'Angelo HM, Fonken LK, Frank MG, Maier SF, Lowry CA. Alzheimer's Disease: Protective Effects of Mycobacterium vaccae, a Soil-Derived Mycobacterium with Anti-Inflammatory and Anti-Tubercular Properties, on the Proteomic Profiles of Plasma and Cerebrospinal Fluid in Rats. J Alzheimers Dis 2020; 78:965-987. [PMID: 33074227 DOI: 10.3233/jad-200568] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is an inflammatory neurodegenerative disease that may be associated with prior bacterial infections. Microbial "old friends" can suppress exaggerated inflammation in response to disease-causing infections or increase clearance of pathogens such as Mycobacterium tuberculosis, which causes tuberculosis (TB). One such "old friend" is Mycobacterium vaccae NCTC 11659, a soil-derived bacterium that has been proposed either as a vaccine for prevention of TB, or as immunotherapy for the treatment of TB when used alongside first line anti-TB drug treatment. OBJECTIVE The goal of this study was to use a hypothesis generating approach to explore the effects of M. vaccae on physiological changes in the plasma and cerebrospinal fluid (CSF). METHODS Liquid chromatography-tandem mass spectrometry-based proteomics were performed in plasma and CSF of adult male rats after immunization with a heat-killed preparation of M. vaccae NCTC 11659 or borate-buffered saline vehicle. Gene enrichment analysis and analysis of protein-protein interactions were performed to integrate physiological network changes in plasma and CSF. We used RT-qPCR to assess immune and metabolic gene expression changes in the hippocampus. RESULTS In both plasma and CSF, immunization with M. vaccae increased proteins associated with immune activation and downregulated proteins corresponding to lipid (including phospholipid and cholesterol) metabolism. Immunization with M. vaccae also increased hippocampal expression of interleukin-4 (IL-4) mRNA, implicating anti-inflammatory effects in the central nervous system. CONCLUSION M. vaccae alters host immune activity and lipid metabolism. These data are consistent with the hypothesis that microbe-host interactions may protect against possible infection-induced, inflammation-related cognitive impairments.
Collapse
Affiliation(s)
- Kelsey M Loupy
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Thomas Lee
- Central Analytical Laboratory and Mass Spectrometry Facility, Department of Biochemistry, University of Colorado Boulder, Boulder, CO, USA
| | - Cristian A Zambrano
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Ahmed I Elsayed
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Heather M D'Angelo
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - Laura K Fonken
- Division of Pharmacology and Toxicology, University of Texas at Austin, Austin, TX, USA
| | - Matthew G Frank
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, USA.,Center for Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - Steven F Maier
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, USA.,Center for Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - Christopher A Lowry
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA.,Center for Neuroscience, University of Colorado Boulder, Boulder, CO, USA.,Center for Microbial Exploration, University of Colorado Boulder, Boulder, CO, USA.,Department of Physical Medicine and Rehabilitation and Center for Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.,Veterans Health Administration, Rocky Mountain Mental Illness Research Education and Clinical Center (MIRECC), Rocky Mountain Regional Veterans Affairs Medical Center (RMRVAMC), Aurora, CO, USA.,Military and Veteran Microbiome: Consortium for Research and Education (MVM-CoRE), Aurora, CO, USA.,Senior Fellow, inVIVO Planetary Health, of the Worldwide Universities Network (WUN), West New York, NJ, USA
| |
Collapse
|
31
|
Baka R, Eckersall D, Horvatic A, Gelemanovic A, Mrljak V, McLaughlin M, Athanasiou LV, Papaioannou N, Stylianaki I, Hanh HQ, Chadwick CC, Polizopoulou Z. Quantitative proteomics of cerebrospinal fluid using tandem mass tags in dogs with recurrent epileptic seizures. J Proteomics 2020; 231:103997. [PMID: 33011347 DOI: 10.1016/j.jprot.2020.103997] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 08/12/2020] [Accepted: 09/27/2020] [Indexed: 02/06/2023]
Abstract
This prospective study included four dog groups (group A: healthy dogs, groups B: dogs with idiopathic epilepsy under antiepileptic medication (AEM), C: idiopathic epilepsy dogs without AEM administration, D: dogs with structural epilepsy). The purpose of the study was to compare the proteomic profile among the four groups. Samples were analyzed by a quantitative Tandem Mass Tags approach using a Q-Exactive-Plus mass-spectrometer. Identification and relative quantification were performed using Proteome Discoverer, and data were analyzed using R. Gene ontology terms were analyzed based on Canis lupus familiaris database. Data are available via ProteomeXchange with identifier PXD018893. Eighteen proteins were statistically significant among the four groups (P < 0.05). MMP2 and EFEMP2 appeared down-regulated whereas HP and APO-A1 were up-regulated (groups B, D). CLEC3B and PEBP4 were up-regulated whereas APO-A1 was down-regulated (group C). IGLL1 was down-regulated (groups B, C) and up-regulated (group D). EFEMP2 was the only protein detected among the four groups and PEBP4 was significantly different among the epileptic dogs. Western blot and SPARCL immunoassay were used to quantify HP abundance change, validating proteomic analysis. Both, showed good correlation with HP levels identified through proteomic analysis (r = 0.712 and r = 0.703, respectively). SIGNIFICANCE: The proteomic analysis from CSF of dogs with epileptic seizures could reflect that MMP2, HP and APO-A1 may contribute to a blood-brain barrier disruption through the seizure-induced inflammatory process in the brain. MMP2 change may indicate the activation of protective mechanisms within the brain tissue. Antiepileptic medication could influence several cellular responses and alter the CSF proteome composition.
Collapse
Affiliation(s)
- Rania Baka
- Diagnostic Laboratory, Faculty of Veterinary Medicine, School of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece.
| | - David Eckersall
- Institute of Biodiversity, Animal Health & Comparative Medicine and School of Veterinary Medicine, College of Medicine, Veterinary Medicine and Life Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - Anita Horvatic
- VetMedZg Laboratory, Faculty of Veterinary Medicine, University of Zagreb, Zagreb, Croatia
| | | | - Vladimir Mrljak
- VetMedZg Laboratory, Faculty of Veterinary Medicine, University of Zagreb, Zagreb, Croatia
| | - Mark McLaughlin
- Institute of Biodiversity, Animal Health & Comparative Medicine and School of Veterinary Medicine, College of Medicine, Veterinary Medicine and Life Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - Labrini V Athanasiou
- Department of Medicine, Faculty of Veterinary Medicine, University of Thessaly, Karditsa, Greece
| | - Nikolaos Papaioannou
- Department of Pathology, Faculty of Veterinary Medicine, School of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Ioanna Stylianaki
- Department of Pathology, Faculty of Veterinary Medicine, School of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Han Quang Hanh
- Institute of Biodiversity, Animal Health & Comparative Medicine and School of Veterinary Medicine, College of Medicine, Veterinary Medicine and Life Sciences, University of Glasgow, Glasgow G61 1QH, UK; Faculty of Animal Science, Vietnam National University of Agriculture, Hanoi, Viet Nam
| | | | - Zoe Polizopoulou
- Diagnostic Laboratory, Faculty of Veterinary Medicine, School of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
32
|
Hu H, Tan L, Bi YL, Xu W, Tan L, Shen XN, Hou XH, Ma YH, Dong Q, Yu JT. Association of serum Apolipoprotein B with cerebrospinal fluid biomarkers of Alzheimer's pathology. Ann Clin Transl Neurol 2020; 7:1766-1778. [PMID: 32910550 PMCID: PMC7545610 DOI: 10.1002/acn3.51153] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/23/2020] [Accepted: 07/16/2020] [Indexed: 12/29/2022] Open
Abstract
Objective To examine whether apolipoprotein B (ApoB), apolipoprotein A‐1 (ApoA1), or their ratio (ApoB/A1) were associated with early changes in cerebrospinal fluid (CSF) biomarkers of Alzheimer’s disease (AD) pathology in elderly adults with subjective cognitive decline (SCD). Methods This study included 507 objective cognitive normal participants from the Chinese Alzheimer’s Biomarker and LifestylE (CABLE) database including 288 cognitive normal participants (CN) and 219 SCD. Multiple linear regression models were used to examine the associations of apolipoproteins with CSF AD biomarkers. Results Compared with control group, SCD participants with significant AD biological characteristics had lower ApoB levels (P = 0.0461). In total participants, lower level of serum ApoB was associated with decreases in CSF Aβ42 (P = 0.0015) and Aβ42/40 (P = 0.0081) as well as increases in CSF p‐tau/Aβ42 (P < 0.0001) and t‐tau/Aβ42 (P = 0.0013), independent of APOEɛ4 status. In further subgroup analysis, these associations were more significant in SCD participants (ApoB × Diagnose: P < 0.05). In addition, lower levels of ApoB were also found associated with increases in p‐tau in the SCD subgroup (P = 0.0263). Furthermore, these protective associations were more significant in the overweight participants (ApoB × weight: P < 0.05). Results showed no association between ApoA1 and CSF biomarkers. Interpretation This study is the first to find protective associations of serum ApoB with CSF AD core biomarkers, especially in SCD individuals. It indicated that ApoB may be a potential biomarker for preclinical AD and may play different roles in different stages of AD.
Collapse
Affiliation(s)
- Hao Hu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yan-Lin Bi
- Department of Anesthesiology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Wei Xu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Lin Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Xue-Ning Shen
- Department of Neurology and Institute of Neurology, WHO Collaborating Center for Research and Training in Neurosciences, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiao-He Hou
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Ya-Hui Ma
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Qiang Dong
- Department of Neurology and Institute of Neurology, WHO Collaborating Center for Research and Training in Neurosciences, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, WHO Collaborating Center for Research and Training in Neurosciences, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
33
|
The Role of HDL and HDL Mimetic Peptides as Potential Therapeutics for Alzheimer's Disease. Biomolecules 2020; 10:biom10091276. [PMID: 32899606 PMCID: PMC7563116 DOI: 10.3390/biom10091276] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 08/25/2020] [Accepted: 08/31/2020] [Indexed: 12/11/2022] Open
Abstract
The role of high-density lipoproteins (HDL) in the cardiovascular system has been extensively studied and the cardioprotective effects of HDL are well established. As HDL particles are formed both in the systemic circulation and in the central nervous system, the role of HDL and its associated apolipoproteins in the brain has attracted much research interest in recent years. Alzheimer’s disease (AD) is the most prevalent neurodegenerative disorder and the leading cause of dementia worldwide, for which there currently exists no approved disease modifying treatment. Multiple lines of evidence, including a number of large-scale human clinical studies, have shown a robust connection between HDL levels and AD. Low levels of HDL are associated with increased risk and severity of AD, whereas high levels of HDL are correlated with superior cognitive function. Although the mechanisms underlying the protective effects of HDL in the brain are not fully understood, many of the functions of HDL, including reverse lipid/cholesterol transport, anti-inflammation/immune modulation, anti-oxidation, microvessel endothelial protection, and proteopathy modification, are thought to be critical for its beneficial effects. This review describes the current evidence for the role of HDL in AD and the potential of using small peptides mimicking HDL or its associated apolipoproteins (HDL-mimetic peptides) as therapeutics to treat AD.
Collapse
|
34
|
Swaminathan SK, Zhou AL, Ahlschwede KM, Curran GL, Lowe VJ, Li L, Kandimalla KK. High-Density Lipoprotein Mimetic Peptide 4F Efficiently Crosses the Blood-Brain Barrier and Modulates Amyloid- β Distribution between Brain and Plasma. J Pharmacol Exp Ther 2020; 375:308-316. [PMID: 32778535 DOI: 10.1124/jpet.120.265876] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 07/24/2020] [Indexed: 12/24/2022] Open
Abstract
Treatments to elevate high-density lipoprotein (HDL) levels in plasma have decreased cerebrovascular amyloid -β (Aβ) deposition and mitigated cognitive decline in Alzheimer disease (AD) transgenic mice. Since the major protein component of HDL particles, apolipoprotein A-I (ApoA-I), has very low permeability at the blood-brain barrier (BBB), we investigated 4F, an 18-amino-acid ApoA-I/HDL mimetic peptide, as a therapeutic alternative. Specifically, we examined the BBB permeability of 4F and its effects on [125I]Aβ trafficking from brain to blood and from blood to brain. After systemic injection in mice, the BBB permeability of [125I]4F, estimated as the permeability-surface area (PS) product, ranged between 2 and 5 × 10-6 ml/g per second in various brain regions. The PS products of [125I]4F were ∼1000-fold higher compared with those determined for [125I]ApoA-I. Moreover, systemic infusion with 4F increased the brain efflux of intracerebrally injected [125I]Aβ42. Conversely, 4F infusion decreased the brain influx of systemically injected [125I]Aβ42. Interestingly, 4F did not significantly alter the brain influx of [125I]Aβ40. To corroborate the in vivo findings, we evaluated the effects of 4F on [125I]Aβ42 transcytosis across polarized human BBB endothelial cell (hCMEC/D3) monolayers. Treatment with 4F increased the abluminal-to-luminal flux and decreased the luminal-to-abluminal flux of [125I]Aβ42 across the hCMEC/D3 monolayers. Additionally, 4F decreased the endothelial accumulation of fluorescein-labeled Aβ42 in the hCMEC/D3 monolayers. These findings provide a mechanistic interpretation for the reductions in brain Aβ burden reported in AD mice after oral 4F administration, which represents a novel strategy for treating AD and cerebral amyloid angiopathy. SIGNIFICANCE STATEMENT: The brain permeability of the ApoA-I mimetic peptide 4F was estimated to be ∼1000-fold greater than ApoA-I after systemic injection of radiolabeled peptide/protein in mice. Further, 4F treatment increased the brain efflux of amyloid -β and also decreased its brain influx, as evaluated in mice and in blood-brain barrier cell monolayers. Thus, 4F represents a potential therapeutic strategy to mitigate brain amyloid accumulation in cerebral amyloid angiopathy and Alzheimer disease.
Collapse
Affiliation(s)
- Suresh K Swaminathan
- Department of Pharmaceutics and Brain Barriers Research Center (S.K.S., A.L.Z., K.M.A., K.K.K.) and Department of Experimental and Clinical Pharmacology (L.L.), University of Minnesota, College of Pharmacy, Minneapolis, Minnesota; Department of Pharmaceutical Sciences, College of Pharmacy, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois (K.M.A.); and Departments of Radiology (G.L.C., V.J.L.) and Neurology (G.L.C.), Mayo Clinic, College of Medicine, Rochester, Minnesota
| | - Andrew L Zhou
- Department of Pharmaceutics and Brain Barriers Research Center (S.K.S., A.L.Z., K.M.A., K.K.K.) and Department of Experimental and Clinical Pharmacology (L.L.), University of Minnesota, College of Pharmacy, Minneapolis, Minnesota; Department of Pharmaceutical Sciences, College of Pharmacy, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois (K.M.A.); and Departments of Radiology (G.L.C., V.J.L.) and Neurology (G.L.C.), Mayo Clinic, College of Medicine, Rochester, Minnesota
| | - Kristen M Ahlschwede
- Department of Pharmaceutics and Brain Barriers Research Center (S.K.S., A.L.Z., K.M.A., K.K.K.) and Department of Experimental and Clinical Pharmacology (L.L.), University of Minnesota, College of Pharmacy, Minneapolis, Minnesota; Department of Pharmaceutical Sciences, College of Pharmacy, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois (K.M.A.); and Departments of Radiology (G.L.C., V.J.L.) and Neurology (G.L.C.), Mayo Clinic, College of Medicine, Rochester, Minnesota
| | - Geoffry L Curran
- Department of Pharmaceutics and Brain Barriers Research Center (S.K.S., A.L.Z., K.M.A., K.K.K.) and Department of Experimental and Clinical Pharmacology (L.L.), University of Minnesota, College of Pharmacy, Minneapolis, Minnesota; Department of Pharmaceutical Sciences, College of Pharmacy, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois (K.M.A.); and Departments of Radiology (G.L.C., V.J.L.) and Neurology (G.L.C.), Mayo Clinic, College of Medicine, Rochester, Minnesota
| | - Val J Lowe
- Department of Pharmaceutics and Brain Barriers Research Center (S.K.S., A.L.Z., K.M.A., K.K.K.) and Department of Experimental and Clinical Pharmacology (L.L.), University of Minnesota, College of Pharmacy, Minneapolis, Minnesota; Department of Pharmaceutical Sciences, College of Pharmacy, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois (K.M.A.); and Departments of Radiology (G.L.C., V.J.L.) and Neurology (G.L.C.), Mayo Clinic, College of Medicine, Rochester, Minnesota
| | - Ling Li
- Department of Pharmaceutics and Brain Barriers Research Center (S.K.S., A.L.Z., K.M.A., K.K.K.) and Department of Experimental and Clinical Pharmacology (L.L.), University of Minnesota, College of Pharmacy, Minneapolis, Minnesota; Department of Pharmaceutical Sciences, College of Pharmacy, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois (K.M.A.); and Departments of Radiology (G.L.C., V.J.L.) and Neurology (G.L.C.), Mayo Clinic, College of Medicine, Rochester, Minnesota
| | - Karunya K Kandimalla
- Department of Pharmaceutics and Brain Barriers Research Center (S.K.S., A.L.Z., K.M.A., K.K.K.) and Department of Experimental and Clinical Pharmacology (L.L.), University of Minnesota, College of Pharmacy, Minneapolis, Minnesota; Department of Pharmaceutical Sciences, College of Pharmacy, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois (K.M.A.); and Departments of Radiology (G.L.C., V.J.L.) and Neurology (G.L.C.), Mayo Clinic, College of Medicine, Rochester, Minnesota
| |
Collapse
|
35
|
Rehiman SH, Lim SM, Neoh CF, Majeed ABA, Chin AV, Tan MP, Kamaruzzaman SB, Ramasamy K. Proteomics as a reliable approach for discovery of blood-based Alzheimer's disease biomarkers: A systematic review and meta-analysis. Ageing Res Rev 2020; 60:101066. [PMID: 32294542 DOI: 10.1016/j.arr.2020.101066] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 04/03/2020] [Accepted: 04/03/2020] [Indexed: 02/08/2023]
Abstract
In order to gauge the impact of proteomics in discovery of Alzheimer's disease (AD) blood-based biomarkers, this study had systematically reviewed articles published between 1984-2019. Articles that fulfilled the inclusion criteria were assessed for risk of bias. A meta-analysis was performed for replicable candidate biomarkers (CB). Of the 1651 articles that were identified, 17 case-control and two cohort studies, as well as three combined case-control and longitudinal designs were shortlisted. A total of 207 AD and mild cognitive impairment (MCI) CB were discovered, with 48 reported in >2 studies. This review highlights six CB, namely alpha-2-macroglobulin (α2M)ps, pancreatic polypeptide (PP)ps, apolipoprotein A-1 (ApoA-1)ps, afaminp, insulin growth factor binding protein-2 (IGFBP-2)ps and fibrinogen-γ-chainp, all of which exhibited consistent pattern of regulation in >three independent cohorts. They are involved in AD pathogenesis via amyloid-beta (Aβ), neurofibrillary tangles, diabetes and cardiovascular diseases (CVD). Meta-analysis indicated that ApoA-1ps was significantly downregulated in AD (SMD = -1.52, 95% CI: -1.89, -1.16, p < 0.00001), with low inter-study heterogeneity (I2 = 0%, p = 0.59). α2Mps was significantly upregulated in AD (SMD = 0.83, 95% CI: 0.05, 1.62, p = 0.04), with moderate inter-study heterogeneity (I2 = 41%, p = 0.19). Both CB are involved in Aβ formation. These findings provide important insights into blood-based AD biomarkers discovery via proteomics.
Collapse
|
36
|
Chiaradia E, Miller I. In slow pace towards the proteome of equine body fluids. J Proteomics 2020; 225:103880. [PMID: 32569818 DOI: 10.1016/j.jprot.2020.103880] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/12/2020] [Accepted: 06/16/2020] [Indexed: 12/14/2022]
Abstract
Equine medicine represents a relevant field of veterinary science and the horse industry generates a significant economic impact. Horses can be involved in different sport disciplines, meat production, work and recreational purposes. Horses are also important for human health as they can be used as animal models for studying human diseases and in equine-assisted therapy. This review summarizes the data related to body fluids such as plasma/serum, urine, cerebrospinal fluid, synovial fluid, saliva, bronchoalveolar lavage fluid and peritoneal fluid obtained using proteomic analysis. Horse body fluid proteome analysis under various physiological and pathological conditions is a useful method for identifying new biomarkers for horse diseases which are still difficult to diagnose, but with serious consequences on equine health and welfare. The findings reported here reveal that further proteomic studies on equine body fluids collected from diseased animals are required. SIGNIFICANCE: Body fluids are sources of potential protein biomarkers for diagnosis and therapeutic target identification. Indeed, they contain proteins that play a crucial role in cell functions and whose presence or relative abundance are indicative of the health status of tissues/organs. The review reports the data on the equine body fluids obtained using proteomic analysis, including those which are commonly used to obtain a correct diagnosis and prognosis of horse diseases which still pose a significant challenge. For equine medicine, new biomarkers are needed to formulate early diagnosis and to distinguish among diseases with similar clinical signs.
Collapse
Affiliation(s)
- Elisabetta Chiaradia
- Laboratory of proteomics, Dipartimento di Medicina Veterinaria, Università degli Studi di Perugia, via San Costanzo, 4, 06126 Perugia, Italy.
| | - Ingrid Miller
- Institut für Medizinische Biochemie, Veterinärmedizinische Universität Wien, Veterinärplatz 1, A-1210 Wien, Austria.
| |
Collapse
|
37
|
Cao W, Zheng D, Wang G, Zhang J, Ge S, Singh M, Wang H, Song M, Li D, Wang W, Xu X, Wang Y. Modelling biological age based on plasma peptides in Han Chinese adults. Aging (Albany NY) 2020; 12:10676-10686. [PMID: 32501290 PMCID: PMC7346055 DOI: 10.18632/aging.103286] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 04/27/2020] [Indexed: 12/18/2022]
Abstract
Age-related disease burdens increased over time, and whether plasma peptides can be used to accurately predict age in order to explain the variation in biological indicators remains inadequately understood. Here we first developed a biological age model based on plasma peptides in 1890 Chinese Han adults. Based on mass spectrometry, 84 peptides were detected with masses in the range of 0.6-10.0 kDa, and 13 of these peptides were identified as known amino acid sequences. Five of these thirteen plasma peptides, including fragments of apolipoprotein A-I (m/z 2883.99), fibrinogen alpha chain (m/z 3060.13), complement C3 (m/z 2190.59), complement C4-A (m/z 1898.21), and breast cancer type 2 susceptibility protein (m/z 1607.84) were finally included in the final model by performing a multivariate linear regression with stepwise selection. This biological age model accounted for 72.3% of the variation in chronological age. Furthermore, the linear correlation between the actual age and biological age was 0.851 (95% confidence interval: 0.836-0.864) and 0.842 (95% confidence interval: 0.810-0.869) in the training and validation sets, respectively. The biological age based on plasma peptides has potential positive effects on primary prevention, and its biological meaning warrants further investigation.
Collapse
Affiliation(s)
- Weijie Cao
- Department of Epidemiology and Health Statistics, School of Public Health, Beijing Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing 100069, China
| | - Deqiang Zheng
- Department of Epidemiology and Health Statistics, School of Public Health, Beijing Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing 100069, China
| | - Guohua Wang
- The Second Affiliated Hospital of Shandong First Medical University, Tai'an 271000, China
| | - Jie Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Beijing Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing 100069, China
| | - Siqi Ge
- Beijing Neurosurgical Institute, Beijing 100070, China
| | - Manjot Singh
- School of Medical and Health Sciences, Edith Cowan University, Perth 6027, Australia
| | - Hao Wang
- Department of Epidemiology and Health Statistics, School of Public Health, Beijing Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing 100069, China.,School of Medical and Health Sciences, Edith Cowan University, Perth 6027, Australia
| | - Manshu Song
- School of Medical and Health Sciences, Edith Cowan University, Perth 6027, Australia
| | - Dong Li
- School of Public Health, Shandong First Medical University and Academy of Medical Sciences of Shandong Province, Tai'an 271016, China
| | - Wei Wang
- Department of Epidemiology and Health Statistics, School of Public Health, Beijing Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing 100069, China.,School of Medical and Health Sciences, Edith Cowan University, Perth 6027, Australia.,School of Public Health, Shandong First Medical University and Academy of Medical Sciences of Shandong Province, Tai'an 271016, China
| | - Xizhu Xu
- School of Public Health, Shandong First Medical University and Academy of Medical Sciences of Shandong Province, Tai'an 271016, China
| | - Youxin Wang
- Department of Epidemiology and Health Statistics, School of Public Health, Beijing Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing 100069, China.,School of Medical and Health Sciences, Edith Cowan University, Perth 6027, Australia
| |
Collapse
|
38
|
Romani A, Trentini A, van der Flier WM, Bellini T, Zuliani G, Cervellati C, Teunissen CE. Arylesterase Activity of Paraoxonase-1 in Serum and Cerebrospinal Fluid of Patients with Alzheimer's Disease and Vascular Dementia. Antioxidants (Basel) 2020; 9:antiox9050456. [PMID: 32466344 PMCID: PMC7278748 DOI: 10.3390/antiox9050456] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/08/2020] [Accepted: 05/22/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND It has been suggested that circulating Paraoxonase-1 (PON1) and apolipoprotein A1 (APOA1), which closely interacts with the antioxidant enzyme, could be implicated in Alzheimer's disease (AD) and vascular dementia (VaD) development. This study aimed to evaluate PON1 changes in serum and cerebrospinal fluid (CSF) as evidence for its association with AD or VaD. METHODS Serum PON-arylesterase activity was measured in patients with AD, VaD, and CONTROLS distributed in two cohorts: Ferrara cohort (FC: n = 503, age = 74 years) and Amsterdam Dementia cohort (ADC: n = 71, age = 65 years). In the last cohort, CSF PON-arylesterase, CSF β-amyloid1-42, p-tau and t-tau, and imaging biomarkers were also measured. RESULTS AD and VaD patients of FC showed significantly lower levels of serum PON-arylesterase compared to CONTROLS, but this outcome was driven by older subjects (>71 years, p < 0.0001). In the younger ADC, a similar decreasing (but not significant) trend was observed in serum and CSF. Intriguingly, PON-arylesterase per APOA1 correlated with t-tau in AD group (r = -0.485, p = 0.002). CONCLUSION These results suggest that decreased peripheral PON-arylesterase might be a specific feature of older AD/VaD patients. Moreover, we showed that PON-arylesterase/APOA1 is inversely related to neurodegeneration in AD patients, suggesting a prognostic usefulness of this composite parameter.
Collapse
Affiliation(s)
- Arianna Romani
- Department of Morphology, Surgery and Experimental Medicine and LTTA Centre, University of Ferrara, 44121 Ferrara, Italy; (A.R.); (G.Z.); (C.C.)
| | - Alessandro Trentini
- Department of Biomedical and Specialist Surgical Sciences, Section of Medical Biochemistry, Molecular Biology and Genetics University of Ferrara, 44121 Ferrara, Italy;
- Correspondence: ; Tel.: +39-053-2455-322
| | - Wiesje M. van der Flier
- Department of Clinical Chemistry, Neurochemistry Lab and Biobank, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (W.M.v.d.F.); (C.E.T.)
- Department of Epidemiology and Biostatistics, Vrije Universiteit Amsterdam, Amsterdam UMC, VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Tiziana Bellini
- Department of Biomedical and Specialist Surgical Sciences, Section of Medical Biochemistry, Molecular Biology and Genetics University of Ferrara, 44121 Ferrara, Italy;
| | - Giovanni Zuliani
- Department of Morphology, Surgery and Experimental Medicine and LTTA Centre, University of Ferrara, 44121 Ferrara, Italy; (A.R.); (G.Z.); (C.C.)
| | - Carlo Cervellati
- Department of Morphology, Surgery and Experimental Medicine and LTTA Centre, University of Ferrara, 44121 Ferrara, Italy; (A.R.); (G.Z.); (C.C.)
| | - Charlotte E. Teunissen
- Department of Clinical Chemistry, Neurochemistry Lab and Biobank, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (W.M.v.d.F.); (C.E.T.)
| |
Collapse
|
39
|
Khoonsari PE, Shevchenko G, Herman S, Remnestål J, Giedraitis V, Brundin R, Degerman Gunnarsson M, Kilander L, Zetterberg H, Nilsson P, Lannfelt L, Ingelsson M, Kultima K. Improved Differential Diagnosis of Alzheimer's Disease by Integrating ELISA and Mass Spectrometry-Based Cerebrospinal Fluid Biomarkers. J Alzheimers Dis 2020; 67:639-651. [PMID: 30614806 PMCID: PMC6398544 DOI: 10.3233/jad-180855] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Background: Alzheimer’s disease (AD) is diagnosed based on a clinical evaluation as well as analyses of classical biomarkers: Aβ42, total tau (t-tau), and phosphorylated tau (p-tau) in cerebrospinal fluid (CSF). Although the sensitivities and specificities of the classical biomarkers are fairly good for detection of AD, there is still a need to develop novel biochemical markers for early detection of AD. Objective: We explored if integration of novel proteins with classical biomarkers in CSF can better discriminate AD from non-AD subjects. Methods: We applied ELISA, mass spectrometry, and multivariate modeling to investigate classical biomarkers and the CSF proteome in subjects (n = 206) with 76 AD patients, 74 mild cognitive impairment (MCI) patients, 11 frontotemporal dementia (FTD) patients, and 45 non-dementia controls. The MCI patients were followed for 4–9 years and 21 of these converted to AD, whereas 53 remained stable. Results: By combining classical CSF biomarkers with twelve novel markers, the area of the ROC curves (AUROCS) of distinguishing AD and MCI/AD converters from non-AD were 93% and 96%, respectively. The FTDs and non-dementia controls were identified versus all other groups with AUROCS of 96% and 87%, respectively. Conclusions: Integration of new and classical CSF biomarkers in a model-based approach can improve the identification of AD, FTD, and non-dementia control subjects.
Collapse
Affiliation(s)
- Payam Emami Khoonsari
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
| | - Ganna Shevchenko
- Department of Chemistry-BMC, Analytical Chemistry, Uppsala University, Uppsala, Sweden
| | - Stephanie Herman
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
| | - Julia Remnestål
- Division of Affinity Proteomics, SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Vilmantas Giedraitis
- Department of Public Health and Caring Sciences/Geriatrics, Uppsala University, Uppsala, Sweden
| | - RoseMarie Brundin
- Department of Public Health and Caring Sciences/Geriatrics, Uppsala University, Uppsala, Sweden
| | | | - Lena Kilander
- Department of Public Health and Caring Sciences/Geriatrics, Uppsala University, Uppsala, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,UK Dementia Research Institute at UCL, London, United Kingdom.,Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, United Kingdom
| | - Peter Nilsson
- Division of Affinity Proteomics, SciLifeLab, Department of Protein Science, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Lars Lannfelt
- Department of Public Health and Caring Sciences/Geriatrics, Uppsala University, Uppsala, Sweden
| | - Martin Ingelsson
- Department of Public Health and Caring Sciences/Geriatrics, Uppsala University, Uppsala, Sweden
| | - Kim Kultima
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
| |
Collapse
|
40
|
Stoye NM, Jung P, Guilherme MDS, Lotz J, Fellgiebel A, Endres K. Apolipoprotein A1 in Cerebrospinal Fluid Is Insufficient to Distinguish Alzheimer's Disease from Other Dementias in a Naturalistic, Clinical Setting. J Alzheimers Dis Rep 2020; 4:15-19. [PMID: 32206754 PMCID: PMC7081088 DOI: 10.3233/adr-190165] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Apolipoprotein A1 (ApoA1) is the major protein component of the high-density lipoprotein and involved in cholesterol transport. Disruption of cholesterol homeostasis has been identified as a contributing factor for Alzheimer’s disease (AD). Moreover, polymorphisms of ApoA1 have been associated with higher risk of disease onset and cognitive decline. Therefore, ApoA1 has been suggested as a biomarker in AD. Here, we tested a small cohort of AD and non-AD dementia patients and measured levels of ApoA1 in cerebrospinal fluid. Our results indicate that ApoA1 might not be applicable to distinguish AD from other forms of dementia.
Collapse
Affiliation(s)
- Nicolai Maximilian Stoye
- Department of Psychiatry and Psychotherapy, University Medical Center Johannes Gutenberg University, Mainz, Germany
| | - Patrick Jung
- Department of Psychiatry and Psychotherapy, University Medical Center Johannes Gutenberg University, Mainz, Germany
| | - Malena Dos Santos Guilherme
- Department of Psychiatry and Psychotherapy, University Medical Center Johannes Gutenberg University, Mainz, Germany
| | - Johannes Lotz
- Institute for Clinical Chemistry and Laboratory Medicine, University Medical Center Johannes Gutenberg University, Mainz, Germany
| | - Andreas Fellgiebel
- Department of Psychiatry and Psychotherapy, University Medical Center Johannes Gutenberg University, Mainz, Germany
| | - Kristina Endres
- Department of Psychiatry and Psychotherapy, University Medical Center Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
41
|
Koch M, DeKosky ST, Goodman M, Sun J, Furtado JD, Fitzpatrick AL, Mackey RH, Cai T, Lopez OL, Kuller LH, Mukamal KJ, Jensen MK. High density lipoprotein and its apolipoprotein-defined subspecies and risk of dementia. J Lipid Res 2019; 61:445-454. [PMID: 31892526 DOI: 10.1194/jlr.p119000473] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 12/19/2019] [Indexed: 11/20/2022] Open
Abstract
Whether HDL is associated with dementia risk is unclear. In addition to apoA1, other apolipoproteins are found in HDL, creating subspecies of HDL that may have distinct metabolic properties. We measured apoA1, apoC3, and apoJ levels in plasma and apoA1 levels in HDL that contains or lacks apoE, apoJ, or apoC3 using a modified sandwich ELISA in a case-cohort study nested within the Ginkgo Evaluation of Memory Study. We included 995 randomly selected participants and 521 participants who developed dementia during a mean of 5.1 years of follow-up. The level of total apoA1 was not significantly related to dementia risk, regardless of the coexistence of apoC3, apoJ, or apoE. Higher levels of total plasma apoC3 were associated with better cognitive function at baseline (difference in Modified Mini-Mental State Examination scores tertile 3 vs. tertile 1: 0.60; 95% CI: 0.23, 0.98) and a lower dementia risk (adjusted hazard ratio tertile 3 vs. tertile 1: 0.73; 95% CI: 0.55, 0.96). Plasma concentrations of apoA1 in HDL and its apolipoprotein-defined subspecies were not associated with cognitive function at baseline or with the risk of dementia during follow-up. Similar studies in other populations are required to better understand the association between apoC3 and Alzheimer's disease pathology.
Collapse
Affiliation(s)
- Manja Koch
- Departments of Nutrition Harvard T.H. Chan School of Public Health, Boston, MA
| | | | - Matthew Goodman
- Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Jiehuan Sun
- Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Jeremy D Furtado
- Departments of Nutrition Harvard T.H. Chan School of Public Health, Boston, MA
| | - Annette L Fitzpatrick
- Departments of Family Medicine, Epidemiology, and Global Health, University of Washington, Seattle, WA
| | - Rachel H Mackey
- Departments of Family Medicine, Epidemiology, and Global Health, University of Washington, Seattle, WA
| | - Tianxi Cai
- Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Oscar L Lopez
- Department of Neurology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA
| | - Lewis H Kuller
- School of Medicine, and Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA
| | - Kenneth J Mukamal
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Majken K Jensen
- Departments of Nutrition Harvard T.H. Chan School of Public Health, Boston, MA.,Department of Medicine, Channing Division of Network Medicine, Harvard Medical School, Brigham and Women's Hospital, Boston, MA
| |
Collapse
|
42
|
Zou S, Zhang J, Chen W. Subtypes Based on Six Apolipoproteins in Non-Demented Elderly Are Associated with Cognitive Decline and Subsequent Tau Accumulation in Cerebrospinal Fluid. J Alzheimers Dis 2019; 72:413-423. [PMID: 31594221 DOI: 10.3233/jad-190314] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Apolipoproteins (APOs) have been implicated in the pathogenesis of Alzheimer's disease (AD). In the present study, we aimed to investigate if patterns of cerebrospinal fluid (CSF) APOs (APOA-I, APOC-III, APOD, APOE, APOH, and APOJ) levels are associated with changes over time in cognition, memory performance, neuroimaging markers, and AD-related pathologies (CSF Aβ42, t-tau, and p-tau) in non-demented older adults. At baseline, a total of 241 non-demented older adults with CSF APOs data was included in the present analysis. Hierarchical agglomerative cluster analysis including the six CSF APOs was carried out. Among non-demented older adults, we identified two clusters. Compare with the first cluster, the second cluster had higher levels of APOs in CSF. Additionally, the second cluster showed a more benign disease course, including slower cognitive decline and slower p-tau accumulation in CSF. Our data highlight the importance of APOs in the pathogenesis of AD.
Collapse
Affiliation(s)
- Shengzhen Zou
- Department of Psychosomatic Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jie Zhang
- Independent Researcher, Hangzhou, China
| | | | - Wei Chen
- Department of Radiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
43
|
Ali JI, Smart CM, Gawryluk JR. Subjective Cognitive Decline and APOE ɛ4: A Systematic Review. J Alzheimers Dis 2019; 65:303-320. [PMID: 30040718 DOI: 10.3233/jad-180248] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Individuals with subjective cognitive decline (SCD) report self-perceived declines in cognitive function but perform within normal limits on standardized tests. However, for some, these self-perceived changes may herald eventual decline to Alzheimer's disease (AD). In light of this, the relationship between SCD and APOE ɛ4, a known genetic risk factor for AD, has garnered interest; however, no systematic review of this literature exists. The current review (n = 36 articles) examined the prevalence of APOE ɛ4 in SCD samples relative to healthy and objectively impaired samples, and summarized APOE ɛ4-related risk of conversion from SCD to AD. Univariate ANOVA indicated that APOE ɛ4 frequency was comparable between healthy control and SCD samples, yet significantly higher in objectively impaired samples (i.e., MCI, AD) relative to either of these groups. Narrative review provided mixed evidence linking coincident APOE ɛ4-positive genotype and SCD to structural neuropathology. Though there was little evidence to suggest that APOE ɛ4 predisposes individuals to developing SCD, both APOE ɛ4 and SCD were found to confer individual and multiplicative risk of conversion to objective cognitive impairment. Combined, it is likely that a relationship between APOE ɛ4, SCD, and AD exists, though its exact nature remains undetermined. A clearer understanding of these relationships is hindered by a lack of standardization in SCD classification and a dearth of longitudinal outcome research. Wide-scale adoption of genetic screening for dementia risk in persons with SCD is considered premature at this time. Ethical considerations and clinical implications of genetic testing for dementia risk are discussed.
Collapse
Affiliation(s)
- Jordan I Ali
- Department of Psychology, University of Victoria, Victoria, BC, Canada.,Institute on Aging & Lifelong Health, University of Victoria, Victoria, BC, Canada
| | - Colette M Smart
- Department of Psychology, University of Victoria, Victoria, BC, Canada.,Institute on Aging & Lifelong Health, University of Victoria, Victoria, BC, Canada
| | - Jodie R Gawryluk
- Department of Psychology, University of Victoria, Victoria, BC, Canada.,Institute on Aging & Lifelong Health, University of Victoria, Victoria, BC, Canada
| |
Collapse
|
44
|
Slot RE, Kester MI, Van Harten AC, Jongbloed W, Bouwman FH, Teunissen CE, Scheltens P, van der Flier WM, Veerhuis R. ApoE and clusterin CSF levels influence associations between APOE genotype and changes in CSF tau, but not CSF Aβ42, levels in non-demented elderly. Neurobiol Aging 2019; 79:101-109. [DOI: 10.1016/j.neurobiolaging.2019.02.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 02/18/2019] [Accepted: 02/22/2019] [Indexed: 01/14/2023]
|
45
|
Dal Magro R, Simonelli S, Cox A, Formicola B, Corti R, Cassina V, Nardo L, Mantegazza F, Salerno D, Grasso G, Deriu MA, Danani A, Calabresi L, Re F. The Extent of Human Apolipoprotein A-I Lipidation Strongly Affects the β-Amyloid Efflux Across the Blood-Brain Barrier in vitro. Front Neurosci 2019; 13:419. [PMID: 31156358 PMCID: PMC6532439 DOI: 10.3389/fnins.2019.00419] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 04/11/2019] [Indexed: 12/20/2022] Open
Abstract
Much evidence suggests a protective role of high-density lipoprotein (HDL) and its major apolipoprotein apoA-I, in Alzheimer's disease (AD). The biogenesis of nascent HDL derived from a first lipidation of apoA-I, which is synthesized by the liver and intestine but not in the brain, in a process mediated by ABCA1. The maturation of nascent HDL in mature spherical HDL is due to a subsequent lipidation step, LCAT-mediated cholesterol esterification, and the change of apoA-I conformation. Therefore, different subclasses of apoA-I-HDL simultaneously exist in the blood circulation. Here, we investigated if and how the lipidation state affects the ability of apoA-I-HDL to target and modulate the cerebral β-amyloid (Aβ) content from the periphery, that is thus far unclear. In particular, different subclasses of HDL, each with different apoA-I lipidation state, were purified from human plasma and their ability to cross the blood-brain barrier (BBB), to interact with Aβ aggregates, and to affect Aβ efflux across the BBB was assessed in vitro using a transwell system. The results showed that discoidal HDL displayed a superior capability to promote Aβ efflux in vitro (9 × 10-5 cm/min), when compared to apoA-I in other lipidation states. In particular, no effect on Aβ efflux was detected when apoA-I was in mature spherical HDL, suggesting that apoA-I conformation, and lipidation could play a role in Aβ clearance from the brain. Finally, when apoA-I folded its structure in discoidal HDL, rather than in spherical ones, it was able to cross the BBB in vitro and strongly destabilize the conformation of Aβ fibrils by decreasing the order of the fibril structure (-24%) and the β-sheet content (-14%). These data suggest that the extent of apoA-I lipidation, and consequently its conformation, may represent crucial features that could exert their protective role in AD pathogenesis.
Collapse
Affiliation(s)
- Roberta Dal Magro
- School of Medicine and Surgery, Nanomedicine Center NANOMIB, University of Milano-Bicocca, Monza, Italy
| | - Sara Simonelli
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Centro Grossi Paoletti, Università degli Studi di Milano, Milan, Italy
| | - Alysia Cox
- School of Medicine and Surgery, Nanomedicine Center NANOMIB, University of Milano-Bicocca, Monza, Italy
| | - Beatrice Formicola
- School of Medicine and Surgery, Nanomedicine Center NANOMIB, University of Milano-Bicocca, Monza, Italy
| | - Roberta Corti
- School of Medicine and Surgery, Nanomedicine Center NANOMIB, University of Milano-Bicocca, Monza, Italy
| | - Valeria Cassina
- School of Medicine and Surgery, Nanomedicine Center NANOMIB, University of Milano-Bicocca, Monza, Italy
| | - Luca Nardo
- School of Medicine and Surgery, Nanomedicine Center NANOMIB, University of Milano-Bicocca, Monza, Italy
| | - Francesco Mantegazza
- School of Medicine and Surgery, Nanomedicine Center NANOMIB, University of Milano-Bicocca, Monza, Italy
| | - Domenico Salerno
- School of Medicine and Surgery, Nanomedicine Center NANOMIB, University of Milano-Bicocca, Monza, Italy
| | - Gianvito Grasso
- Istituto Dalle Molle di Studi sull’Intelligenza Artificiale, Scuola Universitaria Professionale della Svizzera Italiana, Università della Svizzera Italiana, Manno, Switzerland
| | - Marco Agostino Deriu
- Istituto Dalle Molle di Studi sull’Intelligenza Artificiale, Scuola Universitaria Professionale della Svizzera Italiana, Università della Svizzera Italiana, Manno, Switzerland
| | - Andrea Danani
- Istituto Dalle Molle di Studi sull’Intelligenza Artificiale, Scuola Universitaria Professionale della Svizzera Italiana, Università della Svizzera Italiana, Manno, Switzerland
| | - Laura Calabresi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Centro Grossi Paoletti, Università degli Studi di Milano, Milan, Italy
| | - Francesca Re
- School of Medicine and Surgery, Nanomedicine Center NANOMIB, University of Milano-Bicocca, Monza, Italy
| |
Collapse
|
46
|
Contu L, Carare RO, Hawkes CA. Knockout of apolipoprotein A-I decreases parenchymal and vascular β-amyloid pathology in the Tg2576 mouse model of Alzheimer's disease. Neuropathol Appl Neurobiol 2019; 45:698-714. [PMID: 31002190 DOI: 10.1111/nan.12556] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 04/10/2019] [Indexed: 11/30/2022]
Abstract
AIMS Apolipoprotein A-I (apoA-I), the principal apolipoprotein associated with high-density lipoproteins in the periphery, is also found at high concentrations in the cerebrospinal fluid. Previous studies have reported either no impact or vascular-specific effects of apoA-I knockout (KO) on β-amyloid (Aβ) pathology. However, the putative mechanism(s) by which apoA-I may influence Aβ deposition is unknown. METHODS We evaluated the effect of apoA-I deletion on Aβ pathology, Aβ production and clearance from the brain in the Tg2576 mouse model of Alzheimer's disease (AD). RESULTS Contrary to previous reports, deletion of the APOA1 gene significantly reduced concentrations of insoluble Aβ40 and Aβ42 and reduced plaque load in both the parenchyma and blood vessels of apoA-I KO × Tg2576 mice compared to Tg2576 animals. This was not due to decreased Aβ production or alterations in Aβ species. Levels of soluble clusterin/apoJ were significantly higher in neurons of apoA-I KO mice compared to both wildtype (WT) and apoA-I KO × Tg2576 mice. In addition, clearance of Aβ along intramural periarterial drainage pathways was significantly higher in apoA-I KO mice compared to WT animals. CONCLUSION These data suggest that deletion of apoA-I is associated with increased clearance of Aβ and reduced parenchymal and vascular Aβ pathology in the Tg2576 model. These results suggest that peripheral dyslipidaemia can modulate the expression of apolipoproteins in the brain and may influence Aβ clearance and aggregation in AD.
Collapse
Affiliation(s)
- L Contu
- School of Life, Health and Chemical Sciences, STEM Faculty, The Open University, Milton Keynes, UK
| | - R O Carare
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - C A Hawkes
- School of Life, Health and Chemical Sciences, STEM Faculty, The Open University, Milton Keynes, UK
| |
Collapse
|
47
|
Liu S, Suzuki H, Ito H, Korenaga T, Akatsu H, Meno K, Uchida K. Serum levels of proteins involved in amyloid-β clearance are related to cognitive decline and neuroimaging changes in mild cognitive impairment. ALZHEIMER'S & DEMENTIA: DIAGNOSIS, ASSESSMENT & DISEASE MONITORING 2019; 11:85-97. [PMID: 30671532 PMCID: PMC6335589 DOI: 10.1016/j.dadm.2018.11.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Introduction Amyloid-β (Aβ) clearance is important for damage prevention in Alzheimer's disease. We investigated the utility of Aβ clearance proteins as biomarkers for mild cognitive impairment (MCI). Methods Serum apolipoprotein (apo) A-I, compliment protein C3 (C3), transthyretin, and cholesterol levels were measured in 273 subjects, and we analyzed the relationship between these levels and brain atrophy and cerebral blood flow in 63 clinically diagnosed mild cognitive impairment, Alzheimer's disease, and nondemented disease control subjects. Results ApoA-I and transthyretin levels and the active form of C3:native form of C3 ratio achieved an area under the curve of 0.89 (sensitivity: 83%, specificity: 90%) for detecting late mild cognitive impairment. Atrophy was associated with decreased apoA-I and high-density lipoprotein levels. Subjects with reduced cerebral blood flow had lower levels of active form of C3, apoA-I, high-density lipoprotein, and total cholesterol. Low native form of C3 and high active form of C3 levels were found in the hippocampi of patients with Alzheimer's disease. Discussion Aβ clearance proteins in the serum are potential biomarkers for mild cognitive impairment evaluation.
Collapse
Affiliation(s)
- Shan Liu
- Department of Molecular Biological Oncology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Department of Neuropsychiatry, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Tsukuba Industrial Liaison and Cooperative Research Center, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Hideaki Suzuki
- Tsukuba Industrial Liaison and Cooperative Research Center, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Research Division, MCBI. Inc., Ibaraki, Japan
| | - Hitomi Ito
- Tsukuba Industrial Liaison and Cooperative Research Center, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Research Division, MCBI. Inc., Ibaraki, Japan
| | - Tatsumi Korenaga
- Tsukuba Industrial Liaison and Cooperative Research Center, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Research Division, MCBI. Inc., Ibaraki, Japan
| | | | - Kohji Meno
- Tsukuba Industrial Liaison and Cooperative Research Center, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Research Division, MCBI. Inc., Ibaraki, Japan
| | - Kazuhiko Uchida
- Department of Molecular Biological Oncology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Tsukuba Industrial Liaison and Cooperative Research Center, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Corresponding author. Tel.: +81-29-853-3210; Fax: +81-50-3730-7456.
| |
Collapse
|
48
|
Westwood S, Baird AL, Hye A, Ashton NJ, Nevado-Holgado AJ, Anand SN, Liu B, Newby D, Bazenet C, Kiddle SJ, Ward M, Newton B, Desai K, Tan Hehir C, Zanette M, Galimberti D, Parnetti L, Lleó A, Baker S, Narayan VA, van der Flier WM, Scheltens P, Teunissen CE, Visser PJ, Lovestone S. Plasma Protein Biomarkers for the Prediction of CSF Amyloid and Tau and [ 18F]-Flutemetamol PET Scan Result. Front Aging Neurosci 2018; 10:409. [PMID: 30618716 PMCID: PMC6297196 DOI: 10.3389/fnagi.2018.00409] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 11/28/2018] [Indexed: 01/01/2023] Open
Abstract
Background: Blood biomarkers may aid in recruitment to clinical trials of Alzheimer's disease (AD) modifying therapeutics by triaging potential trials participants for amyloid positron emission tomography (PET) or cerebrospinal fluid (CSF) Aβ and tau tests. Objective: To discover a plasma proteomic signature associated with CSF and PET measures of AD pathology. Methods: Liquid chromatography-tandem mass spectrometry (LC-MS/MS) based proteomics were performed in plasma from participants with subjective cognitive decline (SCD), mild cognitive impairment (MCI), and AD, recruited to the Amsterdam Dementia Cohort, stratified by CSF Tau/Aβ42 (n = 50). Technical replication and independent validation were performed by immunoassay in plasma from SCD, MCI, and AD participants recruited to the Amsterdam Dementia Cohort with CSF measures (n = 100), MCI participants enrolled in the GE067-005 study with [18F]-Flutemetamol PET amyloid measures (n = 173), and AD, MCI and cognitively healthy participants from the EMIF 500 study with CSF Aβ42 measurements (n = 494). Results: 25 discovery proteins were nominally associated with CSF Tau/Aβ42 (P < 0.05) with associations of ficolin-2 (FCN2), apolipoprotein C-IV and fibrinogen β chain confirmed by immunoassay (P < 0.05). In the GE067-005 cohort, FCN2 was nominally associated with PET amyloid (P < 0.05) replicating the association with CSF Tau/Aβ42. There were nominally significant associations of complement component 3 with PET amyloid, and apolipoprotein(a), apolipoprotein A-I, ceruloplasmin, and PPY with MCI conversion to AD (all P < 0.05). In the EMIF 500 cohort FCN2 was trending toward a significant relationship with CSF Aβ42 (P ≈ 0.05), while both A1AT and clusterin were nominally significantly associated with CSF Aβ42 (both P < 0.05). Conclusion: Associations of plasma proteins with multiple measures of AD pathology and progression are demonstrated. To our knowledge this is the first study to report an association of FCN2 with AD pathology. Further testing of the proteins in larger independent cohorts will be important.
Collapse
Affiliation(s)
- Sarah Westwood
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - Alison L. Baird
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - Abdul Hye
- Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kigndom
- Biomedical Research Unit for Dementia, NIHR Biomedical Research Centre for Mental Health, South London and Maudsley NHS Foundation Trust, London, United Kingdom
| | - Nicholas J. Ashton
- Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kigndom
- Biomedical Research Unit for Dementia, NIHR Biomedical Research Centre for Mental Health, South London and Maudsley NHS Foundation Trust, London, United Kingdom
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | | | - Sneha N. Anand
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - Benjamine Liu
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - Danielle Newby
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - Chantal Bazenet
- Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kigndom
| | - Steven J. Kiddle
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
- MRC Biostatistics Unit, Cambridge Biomedical Campus, Cambridge Institute of Public Health, University of Cambridge, Cambridge, United Kingdom
| | - Malcolm Ward
- Proteomics Facility, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
| | - Ben Newton
- GE Healthcare Life Sciences Core Imaging, London, United Kingdom
| | - Keyur Desai
- Biosciences, GE Global Research, Niskayuna, NY, United States
| | | | - Michelle Zanette
- GE Healthcare Life Sciences Core Imaging, Marlborough, MA, United States
| | - Daniela Galimberti
- Neurodegenerative Diseases Unit, Centro Dino Ferrari, University of Milan, Milan, Italy
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Lucilla Parnetti
- Center for Memory Disorders and Laboratory of Clinical Neurochemistry, Neurology Clinic, University of Perugia, Perugia, Italy
| | - Alberto Lleó
- Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Susan Baker
- Janssen Neuroscience Research & Development, Titusville, NJ, United States
| | - Vaibhav A. Narayan
- Janssen Neuroscience Research & Development, Titusville, NJ, United States
| | - Wiesje M. van der Flier
- Department of Neurology, Alzheimer Centre, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, Netherlands
- Department of Epidemiology and Biostatistics, VU University Medical Center, Amsterdam, Netherlands
| | - Philip Scheltens
- Department of Neurology, Alzheimer Centre, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, Netherlands
| | - Charlotte E. Teunissen
- Department of Clinical Chemistry, Neurochemistry Lab and Biobank, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, Netherlands
| | - Pieter Jelle Visser
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Simon Lovestone
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
49
|
Lin CY, Sheu JJ, Tsai IS, Wang ST, Yang LY, Hsu IU, Chang HW, Lee HM, Kao SH, Lee CK, Chen CH, Lin YF. Elevated IgM against Nε-(Carboxyethyl)lysine-modified Apolipoprotein A1 peptide 141–147 in Taiwanese with Alzheimer's disease. Clin Biochem 2018; 56:75-82. [DOI: 10.1016/j.clinbiochem.2018.04.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 03/02/2018] [Accepted: 04/10/2018] [Indexed: 12/21/2022]
|
50
|
Dogan S, Paulus M, Forov Y, Weis C, Kampmann M, Cewe C, Kiesel I, Degen P, Salmen P, Rehage H, Tolan M. Human Apolipoprotein A1 at Solid/Liquid and Liquid/Gas Interfaces. J Phys Chem B 2018; 122:3953-3960. [PMID: 29488751 DOI: 10.1021/acs.jpcb.7b12481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
An X-ray reflectivity study on the adsorption behavior of human apolipoprotein A1 (apoA1) at hydrophilic and hydrophobic interfaces is presented. It is shown that the protein interacts via electrostatic and hydrophobic interactions with the interfaces, resulting in the absorption of the protein. pH dependent measurements at the solid/liquid interface between silicon dioxide and aqueous protein solution show that in a small pH range between pH 4 and 6, adsorption is increased due to electrostatic attraction. Here, the native shape of the protein seems to be conserved. In contrast, the adsorption at the liquid/gas interface is mainly driven by hydrophobic effects, presumably by extending the hydrophobic regions of the amphipathic helices, and results in a conformational change of the protein during adsorption. However, the addition of differently charged membrane-forming lipids at the liquid/gas interface illustrates the ability of apoA1 to include lipids, resulting in a depletion of the lipids from the interface.
Collapse
|