51
|
McComb M, Krikheli M, Uher T, Browne RW, Srpova B, Oechtering J, Maceski AM, Tyblova M, Jakimovski D, Ramasamy DP, Bergsland N, Krasensky J, Noskova L, Fialova L, Weinstock-Guttman B, Havrdova EK, Vaneckova M, Zivadinov R, Horakova D, Kuhle J, Ramanathan M. Neuroprotective associations of apolipoproteins A-I and A-II with neurofilament levels in early multiple sclerosis. J Clin Lipidol 2020; 14:675-684.e2. [DOI: 10.1016/j.jacl.2020.07.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/24/2020] [Accepted: 07/01/2020] [Indexed: 12/16/2022]
|
52
|
Ng SS, Park JE, Meng W, Chen CP, Kalaria RN, McCarthy NE, Sze SK. Pulsed SILAM Reveals In Vivo Dynamics of Murine Brain Protein Translation. ACS OMEGA 2020; 5:13528-13540. [PMID: 32566817 PMCID: PMC7301365 DOI: 10.1021/acsomega.9b04439] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 05/08/2020] [Indexed: 06/11/2023]
Abstract
Identification of proteins that are synthesized de novo in response to specific microenvironmental cues is critical for understanding molecular mechanisms that underpin vital physiological processes and pathologies. Here, we report that a brief period of SILAM (Stable Isotope Labeling of Mammals) diet enables the determination of biological functions corresponding to actively translating proteins in the mouse brain. Our results demonstrate that the synapse, dendrite, and myelin sheath are highly active neuronal structures that display rapid protein synthesis, producing key mediators of chemical signaling as well as nutrient sensing, lipid metabolism, and amyloid precursor protein processing/stability. Together, these findings confirm that protein metabolic activity varies significantly between brain functional units in vivo. Our data indicate that pulsed SILAM approaches can unravel complex protein expression dynamics in the murine brain and identify active synthetic pathways and associated functions that are likely impaired in neurodegenerative diseases.
Collapse
Affiliation(s)
- Ser Sue Ng
- School
of Biological Sciences, Nanyang Technological
University, 60 Nanyang Drive, 637551 Singapore
| | - Jung Eun Park
- School
of Biological Sciences, Nanyang Technological
University, 60 Nanyang Drive, 637551 Singapore
| | - Wei Meng
- School
of Biological Sciences, Nanyang Technological
University, 60 Nanyang Drive, 637551 Singapore
| | - Christopher P. Chen
- Memory,
Aging and Cognition Centre, National University
Health System, 1E Kent
Ridge Road, 119228 Singapore
- Department
of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Blk MD3, 16 Medical Drive, 117600 Singapore
| | - Raj N. Kalaria
- Institute
of Neuroscience, Campus for Ageing and Vitality, Newcastle University, Newcastle
upon Tyne NE4 5PL, U.K.
| | - Neil E. McCarthy
- Centre
for Immunobiology, The Blizard Institute, Bart’s and The London
School of Medicine and Dentistry, Queen
Mary University of London, 4 Newark St, London E1
2AT, U.K.
| | - Siu Kwan Sze
- School
of Biological Sciences, Nanyang Technological
University, 60 Nanyang Drive, 637551 Singapore
| |
Collapse
|
53
|
The Reissner Fiber Is Highly Dynamic In Vivo and Controls Morphogenesis of the Spine. Curr Biol 2020; 30:2353-2362.e3. [PMID: 32386529 DOI: 10.1016/j.cub.2020.04.015] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 02/29/2020] [Accepted: 04/06/2020] [Indexed: 01/08/2023]
Abstract
Cerebrospinal fluid (CSF) physiology is important for the development and homeostasis of the central nervous system, and its disruption has been linked to scoliosis in zebrafish [1, 2]. Suspended in the CSF is an extracellular structure called the Reissner fiber, which extends from the brain through the central canal of the spinal cord. Zebrafish scospondin-null mutants are unable to assemble a Reissner fiber and fail to form a straight body axis during embryonic development [3]. Here, we describe hypomorphic missense mutations of scospondin, which allow Reissner fiber assembly and extension of a straight axis. However, during larval development, these mutants display progressive Reissner fiber disassembly, which is concomitant with the emergence of axial curvatures and scoliosis in adult animals. Using a scospondin-GFP knockin zebrafish line, we demonstrate several dynamic properties of the Reissner fiber in vivo, including embryonic fiber assembly, the continuous rostral to caudal movement of the fiber within the brain and central canal, and subcommissural organ (SCO)-spondin-GFP protein secretion from the floor plate to merge with the fiber. Finally, we show that disassembly of the Reissner fiber is also associated with the progression of axial curvatures in distinct scoliosis mutant zebrafish models. Together, these data demonstrate a critical role for the Reissner fiber for the maintenance of a straight body axis and spine morphogenesis in adult zebrafish. Our study establishes a framework for future investigations to address the cellular effectors responsible for Reissner-fiber-dependent regulation of axial morphology. VIDEO ABSTRACT.
Collapse
|
54
|
Hui L, Soliman ML, Geiger NH, Miller NM, Afghah Z, Lakpa KL, Chen X, Geiger JD. Acidifying Endolysosomes Prevented Low-Density Lipoprotein-Induced Amyloidogenesis. J Alzheimers Dis 2020; 67:393-410. [PMID: 30594929 DOI: 10.3233/jad-180941] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cholesterol dyshomeostasis has been linked to the pathogenesis of sporadic Alzheimer's disease (AD). In furthering the understanding of mechanisms by which increased levels of circulating cholesterol augments the risk of developing sporadic AD, others and we have reported that low-density lipoprotein (LDL) enters brain parenchyma by disrupting the blood-brain barrier and that endolysosome de-acidification plays a role in LDL-induced amyloidogenesis in neurons. Here, we tested the hypothesis that endolysosome de-acidification was central to amyloid-β (Aβ) generation and that acidifying endolysosomes protects against LDL-induced increases in Aβ levels in neurons. We demonstrated that LDL, but not HDL, de-acidified endolysosomes and increased intraneuronal and secreted levels of Aβ. ML-SA1, an agonist of endolysosome-resident TRPML1 channels, acidified endolysosomes, and TRPML1 knockdown attenuated ML-SA1-induced endolysosome acidification. ML-SA1 blocked LDL-induced increases in intraneuronal and secreted levels of Aβ as well as Aβ accumulation in endolysosomes, prevented BACE1 accumulation in endolysosomes, and decreased BACE1 activity levels. LDL downregulated TRPML1 protein levels, and TRPML1 knockdown worsens LDL-induced increases in Aβ. Our findings suggest that endolysosome acidification by activating TRPML1 may represent a protective strategy against sporadic AD.
Collapse
Affiliation(s)
- Liang Hui
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Mahmoud L Soliman
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Nicholas H Geiger
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Nicole M Miller
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Zahra Afghah
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Koffi L Lakpa
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Xuesong Chen
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Jonathan D Geiger
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| |
Collapse
|
55
|
Flowers SA, Grant OC, Woods RJ, Rebeck GW. O-glycosylation on cerebrospinal fluid and plasma apolipoprotein E differs in the lipid-binding domain. Glycobiology 2020; 30:74-85. [PMID: 31616924 PMCID: PMC7335482 DOI: 10.1093/glycob/cwz084] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 10/01/2019] [Accepted: 10/02/2019] [Indexed: 01/25/2023] Open
Abstract
The O-glycoprotein apolipoprotein E (APOE), the strongest genetic risk factor for Alzheimer's disease, associates with lipoproteins. Cerebrospinal fluid (CSF) APOE binds only high-density lipoproteins (HDLs), while plasma APOE attaches to lipoproteins of diverse sizes with binding fine-tuned by the C-terminal loop. To better understand the O-glycosylation on this critical molecule and differences across tissues, we analyzed the O-glycosylation on APOE isolated from the plasma and CSF of aged individuals. Detailed LC-MS/MS analyses allowed the identification of the glycosite and the attached glycan and site occupancy for all detectable glycosites on APOE and further three-dimensional modeling of physiological glycoforms of APOE. APOE is O-glycosylated at several sites: Thr8, Thr18, Thr194, Ser197, Thr289, Ser290 and Ser296. Plasma APOE held more abundant (20.5%) N-terminal (Thr8) sialylated core 1 (Neu5Acα2-3Galβ1-3GalNAcα1-) glycosylation compared to CSF APOE (0.1%). APOE was hinge domain glycosylated (Thr194 and Ser197) in both CSF (27.3%) and plasma (10.3%). CSF APOE held almost 10-fold more abundant C-terminal (Thr289, Ser290 and Ser296) glycosylation (36.8% of CSF peptide283-299 was glycosylated, 3.8% of plasma peptide283-299), with sialylated and disialylated (Neu5Acα2-3Galβ1-3(Neu5Acα2-6) GalNAcα1-) core 1 structures. Modeling suggested that C-terminal glycosylation, particularly the branched disialylated structure, could interact across domains including the receptor-binding domain. These data, although limited by sample size, suggest that there are tissue-specific APOE glycoforms. Sialylated glycans, previously shown to improve HDL binding, are more abundant on the lipid-binding domain of CSF APOE and reduced in plasma APOE. This indicates that APOE glycosylation may be implicated in lipoprotein-binding flexibility.
Collapse
Affiliation(s)
- Sarah A Flowers
- Department of Neuroscience, Georgetown University, 3970 Reservoir Rd NW, Washington DC 20007, USA
| | - Oliver C Grant
- Biochemistry and Molecular Chemistry, Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Rd, Athens, GA 30602, USA
| | - Robert J Woods
- Biochemistry and Molecular Chemistry, Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Rd, Athens, GA 30602, USA
| | - G William Rebeck
- Department of Neuroscience, Georgetown University, 3970 Reservoir Rd NW, Washington DC 20007, USA
| |
Collapse
|
56
|
Crivelli SM, Giovagnoni C, Visseren L, Scheithauer AL, de Wit N, den Hoedt S, Losen M, Mulder MT, Walter J, de Vries HE, Bieberich E, Martinez-Martinez P. Sphingolipids in Alzheimer's disease, how can we target them? Adv Drug Deliv Rev 2020; 159:214-231. [PMID: 31911096 DOI: 10.1016/j.addr.2019.12.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 10/09/2019] [Accepted: 12/31/2019] [Indexed: 01/06/2023]
Abstract
Altered levels of sphingolipids and their metabolites in the brain, and the related downstream effects on neuronal homeostasis and the immune system, provide a framework for understanding mechanisms in neurodegenerative disorders and for developing new intervention strategies. In this review we will discuss: the metabolites of sphingolipids that function as second messengers; and functional aberrations of the pathway resulting in Alzheimer's disease (AD) pathophysiology. Focusing on the central product of the sphingolipid pathway ceramide, we describ approaches to pharmacologically decrease ceramide levels in the brain and we argue on how the sphingolipid pathway may represent a new framework for developing novel intervention strategies in AD. We also highlight the possible use of clinical and non-clinical drugs to modulate the sphingolipid pathway and sphingolipid-related biological cascades.
Collapse
|
57
|
APOE in the normal brain. Neurobiol Dis 2020; 136:104724. [PMID: 31911114 DOI: 10.1016/j.nbd.2019.104724] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 12/19/2019] [Accepted: 12/31/2019] [Indexed: 12/25/2022] Open
Abstract
The APOE4 protein affects the primary neuropathological markers of Alzheimer's disease (AD): amyloid plaques, neurofibrillary tangles, and gliosis. These interactions have been investigated to understand the strong effect of APOE genotype on risk of AD. However, APOE genotype has strong effects on processes in normal brains, in the absence of the hallmarks of AD. We propose that CNS APOE is involved in processes in the normal brains that in later years apply specifically to processes of AD pathogenesis. We review the differences of the APOE protein found in the CNS compared to the plasma, including post-translational modifications (glycosylation, lipidation, multimer formation), focusing on ways that the common APOE isoforms differ from each other. We also review structural and functional studies of young human brains and control APOE knock-in mouse brains. These approaches demonstrate the effects of APOE genotype on microscopic neuron structure, gross brain structure, and behavior, primarily related to the hippocampal areas. By focusing on the effects of APOE genotype on normal brain function, approaches can be pursued to identify biomarkers of APOE dysfunction, to promote normal functions of the APOE4 isoform, and to prevent the accumulation of the pathologic hallmarks of AD with aging.
Collapse
|
58
|
Abstract
Lipidomics data generated using untargeted mass spectrometry techniques can offer great biological insight to metabolic status and disease diagnoses. As the community's ability to conduct large-scale studies with deep coverage of the lipidome expands, approaches to analyzing untargeted data and extracting biological insight are needed. Currently, the function of most individual lipids are not known; however, meaningful biological information can be extracted. Here, I will describe a step-by-step approach to identify patterns and trends in untargeted mass spectrometry lipidomics data to assist users in extracting information leading to a greater understanding of biological systems.
Collapse
|
59
|
Jin U, Park SJ, Park SM. Cholesterol Metabolism in the Brain and Its Association with Parkinson's Disease. Exp Neurobiol 2019; 28:554-567. [PMID: 31698548 PMCID: PMC6844833 DOI: 10.5607/en.2019.28.5.554] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 10/18/2019] [Accepted: 10/21/2019] [Indexed: 12/11/2022] Open
Abstract
Parkinson’s disease (PD) is the second most progressive neurodegenerative disorder of the aging population after Alzheimer’s disease (AD). Defects in the lysosomal systems and mitochondria have been suspected to cause the pathogenesis of PD. Nevertheless, the pathogenesis of PD remains obscure. Abnormal cholesterol metabolism is linked to numerous disorders, including atherosclerosis. The brain contains the highest level of cholesterol in the body and abnormal cholesterol metabolism links also many neurodegenerative disorders such as AD, PD, Huntington’s disease (HD), and amyotrophic lateral sclerosis (ALS). The blood brain barrier effectively prevents uptake of lipoprotein-bound cholesterol from blood circulation. Accordingly, cholesterol level in the brain is independent from that in peripheral tissues. Because cholesterol metabolism in both peripheral tissue and the brain are quite different, cholesterol metabolism associated with neurodegeneration should be examined separately from that in peripheral tissues. Here, we review and compare cholesterol metabolism in the brain and peripheral tissues. Furthermore, the relationship between alterations in cholesterol metabolism and PD pathogenesis is reviewed.
Collapse
Affiliation(s)
- Uram Jin
- Department of Pharmacology, Ajou University School of Medicine, Suwon 16499, Korea.,Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon 16499, Korea.,Department of Cardiology, Ajou University School of Medicine, Suwon 16499, Korea
| | - Soo Jin Park
- Department of Pharmacology, Ajou University School of Medicine, Suwon 16499, Korea.,Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon 16499, Korea.,Department of Thoracic and Cardiovascular Surgery, Ajou University School of Medicine, Suwon 16499, Korea
| | - Sang Myun Park
- Department of Pharmacology, Ajou University School of Medicine, Suwon 16499, Korea.,Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon 16499, Korea.,BK21 Plus Program, Department of Biomedical Sciences, Ajou University School of Medicine, Suwon 16499, Korea
| |
Collapse
|
60
|
Rey M, Kruse MS, Magrini-Huamán RN, Coirini H. High-Fat Diets and LXRs Expression in Rat Liver and Hypothalamus. Cell Mol Neurobiol 2019; 39:963-974. [PMID: 31161476 DOI: 10.1007/s10571-019-00692-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 05/25/2019] [Indexed: 12/25/2022]
Abstract
Disturbances on lipid metabolism are associated with health disorders. High-fat diets (HFDs) consumption promotes cardiovascular and neurodegenerative diseases where cholesterol plays an important role. Among regulators of this steroid homeostasis, the liver X receptors (LXRs) induce genes that protect cells from cholesterol overload. We previously described how both hypothalamic LXRα and LXRβ are sensitive to a high-fructose diet, suggesting that these receptors trigger responses related to control of energy and food intake. The present work's main objective was to study the effect of different HFDs on LXRs expression (in hypothalamus and liver), and lipid profile. Male rats received control diet (CD), HFD1 (CD + bovine fat (BF)), HFD2 (CD + BF + cholic acid (CA)), HFD3 (CD + BF + cholesterol), or HFD4 (CD + BF + CA + cholesterol) for different time periods. Hypothalamic LXRβ, both hepatic LXRs subtypes, and total cholesterol (TC) raised after 2 weeks of HFDs. Four and 8 weeks of HFD3 and HFD4 increased the LXRs subtypes in both tissues and TC levels. Only HFD4 reduced triglycerides (TG) levels after 2 and 8 weeks. The TC and TG values correlated significantly with LXRs expression only in rats fed with HFD4. These data add relevant information about how diet composition can produce different scales of hypercholesterolemia states accompanied with central and peripheral changes in the LXRs expression.
Collapse
Affiliation(s)
- Mariana Rey
- Laboratorio de Neurobiologia, Instituto de Biologia y Medicina Experimental (IBYME-CONICET), Vuelta de Obligado 2490, C1428ADN, Ciudad Autonoma de Buenos Aires, Buenos Aires, Argentina
| | - María Sol Kruse
- Laboratorio de Neurobiologia, Instituto de Biologia y Medicina Experimental (IBYME-CONICET), Vuelta de Obligado 2490, C1428ADN, Ciudad Autonoma de Buenos Aires, Buenos Aires, Argentina
| | - Rocío Nahimé Magrini-Huamán
- Laboratorio de Neurobiologia, Instituto de Biologia y Medicina Experimental (IBYME-CONICET), Vuelta de Obligado 2490, C1428ADN, Ciudad Autonoma de Buenos Aires, Buenos Aires, Argentina
- Facultad de Ingenieria, Instituto de Biotecnologia, Universidad Nacional de San Juan, Av. Libertador Gral. San Martín 1109, J5400ARL, San Juan, Argentina
- Facultad de Ciencias Medicas, Universidad Catolica de Cuyo, Av. José Ignacio de la Roza 1516, Rivadavia, J5400, San Juan, Argentina
| | - Héctor Coirini
- Laboratorio de Neurobiologia, Instituto de Biologia y Medicina Experimental (IBYME-CONICET), Vuelta de Obligado 2490, C1428ADN, Ciudad Autonoma de Buenos Aires, Buenos Aires, Argentina.
- Facultad de Ciencias Medicas, Universidad Catolica de Cuyo, Av. José Ignacio de la Roza 1516, Rivadavia, J5400, San Juan, Argentina.
- Departamento de Bioquimica Humana, Facultad de Medicina, Universidad de Buenos Aires, Paraguay 2155, 5to Piso, C1121ABG, Ciudad Autonoma de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
61
|
Ohtani R, Nirengi S, Nakamura M, Murase N, Sainouchi M, Kuwata Y, Takata M, Masuda Y, Kotani K, Sakane N. High-Density Lipoprotein Subclasses and Mild Cognitive Impairment: Study of Outcome and aPolipoproteins in Dementia (STOP-Dementia)1. J Alzheimers Dis 2019; 66:289-296. [PMID: 30248050 DOI: 10.3233/jad-180135] [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] [Indexed: 02/01/2023]
Abstract
BACKGROUND High-density lipoprotein (HDL) containing apolipoprotein A-I is associated with the pathogenesis of Alzheimer's disease (AD). HDL particle size is modified in the presence of pathological conditions, while the significance of the HDL particle size remains controversial. OBJECTIVE The aim of this study was to investigate the HDL lipoprotein subclasses in mild cognitive impairment (MCI) and AD. METHODS This cross-sectional study included 20 AD patients, 17 MCI patients, and 17 age-matched controls without cognitive impairment, selected from the database of the Study of Outcome and aPolipoproteins in Dementia (STOP-Dementia) registry. The diagnoses of AD and MCI were performed by expert neurologists according to the Diagnostic and Statistical Manual of Mental Disorders-Fifth Edition criteria. Serum HDL subclasses were measured by electrophoretic separation of lipoproteins using the Lipoprint System. The neutrophil-lymphocyte ratio (NLR), a marker of inflammation, was calculated by dividing the neutrophil count by the lymphocyte count. RESULTS Small-sized HDL particle levels in the MCI group were significantly higher than in the control group, although there was no difference in serum HDL-cholesterol levels between MCI and control groups. NLR in the MCI group was higher than in the control group, but this difference was non-significant (p = 0.09). There was no difference in HDL subclasses or NLR between the AD and control groups. CONCLUSION These findings suggest that HDL subclasses might be associated with the development of MCI.
Collapse
Affiliation(s)
- Ryo Ohtani
- Department of Neurology, National Hospital Organization Kyoto Medical Center, Fukakusa, Kyoto, Japan
| | - Shinsuke Nirengi
- Division of Preventive Medicine, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Fukakusa, Kyoto, Japan
| | - Michikazu Nakamura
- Department of Neurology, National Hospital Organization Kyoto Medical Center, Fukakusa, Kyoto, Japan
| | - Nagako Murase
- Department of Neurology, National Hospital Organization Kyoto Medical Center, Fukakusa, Kyoto, Japan
| | - Makoto Sainouchi
- Department of Neurology, National Hospital Organization Kyoto Medical Center, Fukakusa, Kyoto, Japan
| | - Yasuhiro Kuwata
- Department of Neurology, National Hospital Organization Kyoto Medical Center, Fukakusa, Kyoto, Japan
| | - Masaki Takata
- Department of Neurology, National Hospital Organization Kyoto Medical Center, Fukakusa, Kyoto, Japan
| | - Yuuichi Masuda
- Department of Neurology, National Hospital Organization Kyoto Medical Center, Fukakusa, Kyoto, Japan
| | - Kazuhiko Kotani
- Division of Preventive Medicine, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Fukakusa, Kyoto, Japan.,Division of Community and Family Medicine, Jichi Medical University, Shimotsuke-City, Tochigi, Japan
| | - Naoki Sakane
- Division of Preventive Medicine, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Fukakusa, Kyoto, Japan
| |
Collapse
|
62
|
Picard C, Poirier A, Bélanger S, Labonté A, Auld D, Poirier J. Proprotein convertase subtilisin/kexin type 9 (PCSK9) in Alzheimer's disease: A genetic and proteomic multi-cohort study. PLoS One 2019; 14:e0220254. [PMID: 31437157 PMCID: PMC6705826 DOI: 10.1371/journal.pone.0220254] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 07/11/2019] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a hepatic enzyme that regulates circulating low-density lipoprotein (LDL) cholesterol levels by binding to LDL receptors (LDLR) and promoting their degradation. Although PCSK9 inhibitors were shown to reduce the risk of cardiovascular disease, a warning was issued concerning their possible impact on cognitive functions. In Alzheimer's disease (AD), it is believed that cognitive impairment is associated with cholesterol metabolism alterations, which could involve PCSK9. The main objective of this study is to determine if PCSK9 plays a significant role in the pre-symptomatic phase of the disease when the pathophysiological markers of AD unfolds and, later, when cognitive symptoms emerge. METHODS AND FINDINGS To test if PCSK9 is associated with AD pathology, we measured its expression levels in 65 autopsy confirmed AD brains and 45 age and gender matched controls. Messenger ribonucleic acid (mRNA) were quantified using real-time polymerase chain reaction (RT-PCR) and protein levels were quantified using enzyme-linked immunosorbent assay (ELISA). PCSK9 was elevated in frontal cortices of AD subjects compared to controls, both at the mRNA and protein levels. LDLR protein levels were unchanged in AD frontal cortices, despite and upregulation at the mRNA level. To verify if PCSK9 dysregulation was observable before the onset of AD, we measured its expression in the cerebrospinal fluid (CSF) of 104 "at-risk" subjects and contrasted it with known apolipoproteins levels and specific AD biomarkers using ELISAs. Positive correlations were found between CSF PCSK9 and apolipoprotein E (APOE), apolipoprotein J (APOJ or CLU), apolipoprotein B (APOB), phospho Tau (pTau) and total Tau. To investigate if PCSK9 levels were driven by genetic variants, we conducted an expression quantitative trait loci (eQTL) study using bioinformatic tools and found two polymorphisms in strong association. Further investigation of these variants in two independent cohorts showed a female specific association with AD risk and with CSF Tau levels in cognitively impaired individuals. CONCLUSIONS PCSK9 levels differ between control and AD brains and its protein levels correlate with those of other lipoproteins and AD biomarkers even before the onset of the disease. PCSK9 regulation seems to be under tight genetic control in females only, with specific variants that could predispose to increased AD risk.
Collapse
Affiliation(s)
- Cynthia Picard
- Douglas Mental Health University Institute, Montréal, Québec, Canada
- Centre for the Studies in the Prevention of Alzheimer’s Disease, Montréal, Québec, Canada
- Department of Psychiatry, McGill University, Montréal, Québec, Canada
| | - Alexandre Poirier
- Douglas Mental Health University Institute, Montréal, Québec, Canada
| | | | - Anne Labonté
- Douglas Mental Health University Institute, Montréal, Québec, Canada
- Centre for the Studies in the Prevention of Alzheimer’s Disease, Montréal, Québec, Canada
| | - Daniel Auld
- Génome Québec Innovation Centre, Montréal, Québec, Canada
| | - Judes Poirier
- Douglas Mental Health University Institute, Montréal, Québec, Canada
- Centre for the Studies in the Prevention of Alzheimer’s Disease, Montréal, Québec, Canada
- Department of Psychiatry, McGill University, Montréal, Québec, Canada
| | - on behalf of the PREVENT-AD Research Group
- Douglas Mental Health University Institute, Montréal, Québec, Canada
- Centre for the Studies in the Prevention of Alzheimer’s Disease, Montréal, Québec, Canada
- Department of Psychiatry, McGill University, Montréal, Québec, Canada
| |
Collapse
|
63
|
Chernick D, Ortiz-Valle S, Jeong A, Qu W, Li L. Peripheral versus central nervous system APOE in Alzheimer's disease: Interplay across the blood-brain barrier. Neurosci Lett 2019; 708:134306. [PMID: 31181302 DOI: 10.1016/j.neulet.2019.134306] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 05/28/2019] [Accepted: 05/29/2019] [Indexed: 12/20/2022]
Abstract
The apolipoprotein E (APOE) ε4 allele has been demonstrated as the preeminent genetic risk factor for late onset Alzheimer's disease (AD), which comprises greater than 90% of all AD cases. The discovery of the connection between different APOE genotypes and AD risk in the early 1990s spurred three decades of intense and comprehensive research into the function of APOE in the normal and diseased brain. The importance of APOE in the periphery has been well established, due to its pivotal role in maintaining cholesterol homeostasis and cardiovascular health. The influence of vascular factors on brain function and AD risk has been extensively studied in recent years. As a major apolipoprotein regulating multiple molecular pathways beyond its canonical lipid-related functions in the periphery and the central nervous system, APOE represents a critical link between the two compartments, and may influence AD risk from both sides of the blood-brain barrier. This review discusses recent advances in understanding the different functions of APOE in the periphery and in the brain, and highlights several promising APOE-targeted therapeutic strategies for AD.
Collapse
Affiliation(s)
| | | | - Angela Jeong
- Department of Experimental and Clinical Pharmacology, Minneapolis, MN, United States
| | - Wenhui Qu
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Ling Li
- Departments of Pharmacology, Minneapolis, MN, United States; Department of Experimental and Clinical Pharmacology, Minneapolis, MN, United States; Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, United States.
| |
Collapse
|
64
|
Marchi C, Adorni MP, Caffarra P, Ronda N, Spallazzi M, Barocco F, Galimberti D, Bernini F, Zimetti F. ABCA1- and ABCG1-mediated cholesterol efflux capacity of cerebrospinal fluid is impaired in Alzheimer's disease. J Lipid Res 2019; 60:1449-1456. [PMID: 31167810 DOI: 10.1194/jlr.p091033] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 05/25/2019] [Indexed: 01/18/2023] Open
Abstract
HDL-like particles in human cerebrospinal fluid (CSF) promote the efflux of cholesterol from astrocytes toward the neurons that rely on this supply for their functions. We evaluated whether cell cholesterol efflux capacity of CSF (CSF-CEC) is impaired in Alzheimer's disease (AD) by analyzing AD (n = 37) patients, non-AD dementia (non-AD DEM; n = 16) patients, and control subjects (n = 39). As expected, AD patients showed reduced CSF Aβ 1-42, increased total and phosphorylated tau, and a higher frequency of the apoε4 genotype. ABCA1- and ABCG1-mediated CSF-CEC was markedly reduced in AD (-73% and -33%, respectively) but not in non-AD DEM patients, in which a reduced passive diffusion CEC (-40%) was observed. Non-AD DEM patients displayed lower CSF apoE concentrations (-24%) compared with controls, while apoA-I levels were similar among groups. No differences in CSF-CEC were found by stratifying subjects for apoε4 status. ABCG1 CSF-CEC positively correlated with Aβ 1-42 (r = 0.305, P = 0.025), while ABCA1 CSF-CEC inversely correlated with total and phosphorylated tau (r = -0.348, P = 0.018 and r = -0.294, P = 0.048, respectively). The CSF-CEC impairment and the correlation with the neurobiochemical markers suggest a pathophysiological link between CSF HDL-like particle dysfunction and neurodegeneration in AD.
Collapse
Affiliation(s)
- Cinzia Marchi
- Department of Food and Drug University of Parma, Parma, Italy
| | | | - Paolo Caffarra
- Department of Medicine and Surgery, Section of Neurology University of Parma, Parma, Italy.,Alzheimer Center Briolini Hospital, Gazzaniga, Bergamo, Italy
| | - Nicoletta Ronda
- Department of Food and Drug University of Parma, Parma, Italy
| | - Marco Spallazzi
- Department of Medicine and Surgery, Section of Neurology University of Parma, Parma, Italy
| | - Federica Barocco
- Department of Medicine and Surgery, Section of Neurology University of Parma, Parma, Italy
| | - Daniela Galimberti
- Department of Biomedical, Surgical and Dental Sciences, Dino Ferrari Center, University of Milano, Milano, Italy.,Neurodegenerative Diseases Unit Fondazione Cà Granda, IRCCS Ospedale Maggiore Policlinico, Milano, Italy
| | - Franco Bernini
- Department of Food and Drug University of Parma, Parma, Italy
| | | |
Collapse
|
65
|
Abstract
PURPOSE OF REVIEW We review current knowledge regarding HDL and Alzheimer's disease, focusing on HDL's vasoprotective functions and potential as a biomarker and therapeutic target for the vascular contributions of Alzheimer's disease. RECENT FINDINGS Many epidemiological studies have observed that circulating HDL levels associate with decreased Alzheimer's disease risk. However, it is now understood that the functions of HDL may be more informative than levels of HDL cholesterol (HDL-C). Animal model studies demonstrate that HDL protects against memory deficits, neuroinflammation, and cerebral amyloid angiopathy (CAA). In-vitro studies using state-of-the-art 3D models of the human blood-brain barrier (BBB) confirm that HDL reduces vascular Aβ accumulation and attenuates Aβ-induced endothelial inflammation. Although HDL-based therapeutics have not been tested in clinical trials for Alzheimer's disease , several HDL formulations are in advanced phase clinical trials for coronary artery disease and atherosclerosis and could be leveraged toward Alzheimer's disease . SUMMARY Evidence from human studies, animal models, and bioengineered arteries supports the hypothesis that HDL protects against cerebrovascular dysfunction in Alzheimer's disease. Assays of HDL functions relevant to Alzheimer's disease may be desirable biomarkers of cerebrovascular health. HDL-based therapeutics may also be of interest for Alzheimer's disease, using stand-alone or combination therapy approaches.
Collapse
Affiliation(s)
- Emily B. Button
- Department of Pathology and Laboratory Medicine
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jérôme Robert
- Department of Pathology and Laboratory Medicine
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Tara M. Caffrey
- Department of Pathology and Laboratory Medicine
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jianjia Fan
- Department of Pathology and Laboratory Medicine
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Wenchen Zhao
- Department of Pathology and Laboratory Medicine
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Cheryl L. Wellington
- Department of Pathology and Laboratory Medicine
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
66
|
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
|
67
|
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
|
68
|
Kaiser K, Gyllborg D, Procházka J, Salašová A, Kompaníková P, Molina FL, Laguna-Goya R, Radaszkiewicz T, Harnoš J, Procházková M, Potěšil D, Barker RA, Casado ÁG, Zdráhal Z, Sedláček R, Arenas E, Villaescusa JC, Bryja V. WNT5A is transported via lipoprotein particles in the cerebrospinal fluid to regulate hindbrain morphogenesis. Nat Commun 2019; 10:1498. [PMID: 30940800 PMCID: PMC6445127 DOI: 10.1038/s41467-019-09298-4] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 02/21/2019] [Indexed: 12/11/2022] Open
Abstract
WNTs are lipid-modified proteins that control multiple functions in development and disease via short- and long-range signaling. However, it is unclear how these hydrophobic molecules spread over long distances in the mammalian brain. Here we show that WNT5A is produced by the choroid plexus (ChP) of the developing hindbrain, but not the telencephalon, in both mouse and human. Since the ChP produces and secretes the cerebrospinal fluid (CSF), we examine the presence of WNT5A in the CSF and find that it is associated with lipoprotein particles rather than exosomes. Moreover, since the CSF flows along the apical surface of hindbrain progenitors not expressing Wnt5a, we examined whether deletion of Wnt5a in the ChP controls their function and find that cerebellar morphogenesis is impaired. Our study thus identifies the CSF as a route and lipoprotein particles as a vehicle for long-range transport of biologically active WNT in the central nervous system.
Collapse
Affiliation(s)
- Karol Kaiser
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, 62500, Czech Republic
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 171 77, Sweden
| | - Daniel Gyllborg
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 171 77, Sweden
| | - Jan Procházka
- Czech Centre for Phenogenomics and Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the CAS, v. v. i., Prumyslova 595, Vestec, 252 42, Czech Republic
| | - Alena Salašová
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 171 77, Sweden
- Danish Research Institute of Translational Neuroscience, Department of Biomedicine, Aarhus University, Aarhus, C 8000, Denmark
| | - Petra Kompaníková
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, 62500, Czech Republic
| | - Francisco Lamus Molina
- Departamento de Anatomía y Radiología, Facultad de medicina, Universidad de Valladolid, Ramón y Cajal 5, 47005, Valladolid, Spain
| | - Rocio Laguna-Goya
- John van Geest Centre for Brain Repair and Cambridge Stem Cell Institute, University of Cambridge, Cambridge, CB2 0PY, UK
| | - Tomasz Radaszkiewicz
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, 62500, Czech Republic
| | - Jakub Harnoš
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, 62500, Czech Republic
| | - Michaela Procházková
- Czech Centre for Phenogenomics and Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the CAS, v. v. i., Prumyslova 595, Vestec, 252 42, Czech Republic
| | - David Potěšil
- Central European Institute of Technology, 625 00, Brno, Czech Republic
| | - Roger A Barker
- John van Geest Centre for Brain Repair and Cambridge Stem Cell Institute, University of Cambridge, Cambridge, CB2 0PY, UK
| | - Ángel Gato Casado
- Departamento de Anatomía y Radiología, Facultad de medicina, Universidad de Valladolid, Ramón y Cajal 5, 47005, Valladolid, Spain
| | - Zbyněk Zdráhal
- Central European Institute of Technology, 625 00, Brno, Czech Republic
| | - Radislav Sedláček
- Czech Centre for Phenogenomics and Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the CAS, v. v. i., Prumyslova 595, Vestec, 252 42, Czech Republic
| | - Ernest Arenas
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 171 77, Sweden.
| | - J Carlos Villaescusa
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, 62500, Czech Republic.
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 171 77, Sweden.
- Psychiatric Stem Cell Group, Neurogenetics Unit, Center for Molecular Medicine, Department of Molecular Medicine and Surgery, Karolinska University Hospital, Stockholm, 171 76, Sweden.
| | - Vítězslav Bryja
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, 62500, Czech Republic.
| |
Collapse
|
69
|
Foster EM, Dangla-Valls A, Lovestone S, Ribe EM, Buckley NJ. Clusterin in Alzheimer's Disease: Mechanisms, Genetics, and Lessons From Other Pathologies. Front Neurosci 2019; 13:164. [PMID: 30872998 PMCID: PMC6403191 DOI: 10.3389/fnins.2019.00164] [Citation(s) in RCA: 227] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 02/12/2019] [Indexed: 01/10/2023] Open
Abstract
Clusterin (CLU) or APOJ is a multifunctional glycoprotein that has been implicated in several physiological and pathological states, including Alzheimer's disease (AD). With a prominent extracellular chaperone function, additional roles have been discussed for clusterin, including lipid transport and immune modulation, and it is involved in pathways common to several diseases such as cell death and survival, oxidative stress, and proteotoxic stress. Although clusterin is normally a secreted protein, it has also been found intracellularly under certain stress conditions. Multiple hypotheses have been proposed regarding the origin of intracellular clusterin, including specific biogenic processes leading to alternative transcripts and protein isoforms, but these lines of research are incomplete and contradictory. Current consensus is that intracellular clusterin is most likely to have exited the secretory pathway at some point or to have re-entered the cell after secretion. Clusterin's relationship with amyloid beta (Aβ) has been of great interest to the AD field, including clusterin's apparent role in altering Aβ aggregation and/or clearance. Additionally, clusterin has been more recently identified as a mediator of Aβ toxicity, as evidenced by the neuroprotective effect of CLU knockdown and knockout in rodent and human iPSC-derived neurons. CLU is also the third most significant genetic risk factor for late onset AD and several variants have been identified in CLU. Although the exact contribution of these variants to altered AD risk is unclear, some have been linked to altered CLU expression at both mRNA and protein levels, altered cognitive and memory function, and altered brain structure. The apparent complexity of clusterin's biogenesis, the lack of clarity over the origin of the intracellular clusterin species, and the number of pathophysiological functions attributed to clusterin have all contributed to the challenge of understanding the role of clusterin in AD pathophysiology. Here, we highlight clusterin's relevance to AD by discussing the evidence linking clusterin to AD, as well as drawing parallels on how the role of clusterin in other diseases and pathways may help us understand its biological function(s) in association with AD.
Collapse
Affiliation(s)
| | | | | | | | - Noel J. Buckley
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
70
|
Kielkopf CS, Low JKK, Mok YF, Bhatia S, Palasovski T, Oakley AJ, Whitten AE, Garner B, Brown SHJ. Identification of a novel tetrameric structure for human apolipoprotein-D. J Struct Biol 2018; 203:205-218. [PMID: 29885491 DOI: 10.1016/j.jsb.2018.05.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 05/18/2018] [Accepted: 05/30/2018] [Indexed: 10/14/2022]
Abstract
Apolipoprotein-D is a 25 kDa glycosylated member of the lipocalin family that folds into an eight-stranded β-barrel with a single adjacent α-helix. Apolipoprotein-D specifically binds a range of small hydrophobic ligands such as progesterone and arachidonic acid and has an antioxidant function that is in part due to the reduction of peroxidised lipids by methionine-93. Therefore, apolipoprotein-D plays multiple roles throughout the body and is protective in Alzheimer's disease, where apolipoprotein-D overexpression reduces the amyloid-β burden in Alzheimer's disease mouse models. Oligomerisation is a common feature of lipocalins that can influence ligand binding. The native structure of apolipoprotein-D, however, has not been conclusively defined. Apolipoprotein-D is generally described as a monomeric protein, although it dimerises when reducing peroxidised lipids. Here, we investigated the native structure of apolipoprotein-D derived from plasma, breast cyst fluid (BCF) and cerebrospinal fluid. In plasma and cerebrospinal fluid, apolipoprotein-D was present in high-molecular weight complexes, potentially in association with lipoproteins. In contrast, apolipoprotein-D in BCF formed distinct oligomeric species. We assessed apolipoprotein-D oligomerisation using native apolipoprotein-D purified from BCF and a suite of complementary methods, including multi-angle laser light scattering, analytical ultracentrifugation and small-angle X-ray scattering. Our analyses showed that apolipoprotein-D predominantly forms a ∼95 to ∼100 kDa tetramer. Small-angle X-ray scattering analysis confirmed these findings and provided a structural model for apolipoprotein-D tetramer. These data indicate apolipoprotein-D rarely exists as a free monomer under physiological conditions and provide insights into novel native structures of apolipoprotein-D and into oligomerisation behaviour in the lipocalin family.
Collapse
Affiliation(s)
- Claudia S Kielkopf
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia; School of Biological Sciences, University of Wollongong, Wollongong, NSW, Australia; Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia.
| | - Jason K K Low
- School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, Australia.
| | - Yee-Foong Mok
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, Australia.
| | - Surabhi Bhatia
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia; School of Biological Sciences, University of Wollongong, Wollongong, NSW, Australia.
| | - Tony Palasovski
- Illawarra and Shoalhaven Local Health District (ISLHD), Wollongong, NSW, Australia; Specialist Breast Clinic Sutherland Shire and Wollongong, NSW, Australia; Integrated Specialist Health Care Sutherland Shire, NSW, Australia
| | - Aaron J Oakley
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia; Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia; School of Chemistry, University of Wollongong, Wollongong, NSW, Australia.
| | - Andrew E Whitten
- Australian Nuclear Science and Technology Organisation, Lucas Heights, NSW, Australia.
| | - Brett Garner
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia; School of Biological Sciences, University of Wollongong, Wollongong, NSW, Australia; Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia.
| | - Simon H J Brown
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW, Australia; School of Biological Sciences, University of Wollongong, Wollongong, NSW, Australia; Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia.
| |
Collapse
|
71
|
Pate KM, Kim BJ, Shusta EV, Murphy RM. Transthyretin Mimetics as Anti-β-Amyloid Agents: A Comparison of Peptide and Protein Approaches. ChemMedChem 2018; 13:968-979. [PMID: 29512286 PMCID: PMC5991081 DOI: 10.1002/cmdc.201800031] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 03/05/2018] [Indexed: 12/19/2022]
Abstract
β-Amyloid (Aβ) aggregation is causally linked to neuronal pathology in Alzheimer's disease; therefore, several small molecules, antibodies, and peptides have been tested as anti-Aβ agents. We developed two compounds based on the Aβ-binding domain of transthyretin (TTR): a cyclic peptide cG8 and an engineered protein mTTR, and compared them for therapeutically relevant properties. Both mTTR and cG8 inhibit fibrillogenesis of Aβ, with mTTR inhibiting at a lower concentration than cG8. Both inhibit aggregation of amylin but not of α-synuclein. They both bind more Aβ aggregates than monomer, and neither disaggregates preformed fibrils. cG8 retained more of its activity in the presence of biological materials and was more resistant to proteolysis than mTTR. We examined the effect of mTTR or cG8 on Aβ binding to human neurons. When mTTR was co-incubated with Aβ under oligomer-forming conditions, Aβ morphology was drastically changed and Aβ-cell deposition significantly decreased. In contrast, cG8 did not affect morphology but decreased the amount of Aβ deposited. These results provide guidance for further evolution of TTR-mimetic anti-amyloid agents.
Collapse
Affiliation(s)
- Kayla M Pate
- Department of Chemical and Biological Engineering, University of Wisconsin - Madison, 1415 Engineering Drive, Madison, WI, 53706, USA
| | - Brandon J Kim
- Department of Chemical and Biological Engineering, University of Wisconsin - Madison, 1415 Engineering Drive, Madison, WI, 53706, USA
| | - Eric V Shusta
- Department of Chemical and Biological Engineering, University of Wisconsin - Madison, 1415 Engineering Drive, Madison, WI, 53706, USA
| | - Regina M Murphy
- Department of Chemical and Biological Engineering, University of Wisconsin - Madison, 1415 Engineering Drive, Madison, WI, 53706, USA
| |
Collapse
|
72
|
Muñoz SS, Li H, Ruberu K, Chu Q, Saghatelian A, Ooi L, Garner B. The serine protease HtrA1 contributes to the formation of an extracellular 25-kDa apolipoprotein E fragment that stimulates neuritogenesis. J Biol Chem 2018; 293:4071-4084. [PMID: 29414786 PMCID: PMC5857987 DOI: 10.1074/jbc.ra117.001278] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 01/24/2018] [Indexed: 12/31/2022] Open
Abstract
Apolipoprotein-E (apoE) is a glycoprotein highly expressed in the brain, where it appears to play a role in lipid transport, β-amyloid clearance, and neuronal signaling. ApoE proteolytic fragments are also present in the brain, but the enzymes responsible for apoE fragmentation are unknown, and the biological activity of specific apoE fragments remains to be determined. Here we utilized SK-N-SH neuroblastoma cells differentiated into neurons with all-trans-retinoic acid (ATRA) to study extracellular apoE proteolysis. ApoE fragments were detectable in culture supernatants after 3 days, and their levels were increased for up to 9 days in the presence of ATRA. The concentration of apoE fragments was positively correlated with levels of the neuronal maturation markers (PSD95 and SMI32). The most abundant apoE fragments were 25- and 28-kDa N-terminal fragments that both contained sialylated glycosylation and bound to heparin. We detected apoE fragments only in the extracellular milieu and not in cell lysates, suggesting that an extracellular protease contributes to apoE fragmentation. Of note, siRNA-mediated knockdown of high-temperature requirement serine peptidase A1 (HtrA1) and a specific HtrA1 inhibitor reduced apoE 25-kDa fragment formation by 41 and 86%, respectively. Recombinant 25-kDa fragment apoE and full-length apoE both stimulated neuritogenesis in vitro, increasing neuroblastoma neurite growth by more than 2-fold over a 6-day period. This study provides a cellular model for assessing apoE proteolysis, indicates that HtrA1 regulates apoE 25-kDa fragment formation under physiological conditions, and reveals a new neurotrophic function for the apoE 25-kDa fragment.
Collapse
Affiliation(s)
- Sonia Sanz Muñoz
- From the Illawarra Health and Medical Research Institute and
- the School of Biological Sciences, University of Wollongong, New South Wales 2522, Australia and
| | - Hongyun Li
- From the Illawarra Health and Medical Research Institute and
- the School of Biological Sciences, University of Wollongong, New South Wales 2522, Australia and
| | - Kalani Ruberu
- From the Illawarra Health and Medical Research Institute and
- the School of Biological Sciences, University of Wollongong, New South Wales 2522, Australia and
| | - Qian Chu
- the Clayton Foundation Laboratories for Peptide Biology, Salk Institute for Biological Studies, La Jolla, California 92037
| | - Alan Saghatelian
- the Clayton Foundation Laboratories for Peptide Biology, Salk Institute for Biological Studies, La Jolla, California 92037
| | - Lezanne Ooi
- From the Illawarra Health and Medical Research Institute and
- the School of Biological Sciences, University of Wollongong, New South Wales 2522, Australia and
| | - Brett Garner
- From the Illawarra Health and Medical Research Institute and
- the School of Biological Sciences, University of Wollongong, New South Wales 2522, Australia and
| |
Collapse
|
73
|
Rosenberg JB, Kaplitt MG, De BP, Chen A, Flagiello T, Salami C, Pey E, Zhao L, Ricart Arbona RJ, Monette S, Dyke JP, Ballon DJ, Kaminsky SM, Sondhi D, Petsko GA, Paul SM, Crystal RG. AAVrh.10-Mediated APOE2 Central Nervous System Gene Therapy for APOE4-Associated Alzheimer's Disease. HUM GENE THER CL DEV 2018; 29:24-47. [PMID: 29409358 DOI: 10.1089/humc.2017.231] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive degenerative neurological disorder affecting nearly one in nine elderly people in the United States. Population studies have shown that an inheritance of the apolipoprotein E (APOE) variant APOE4 allele increases the risk of developing AD, whereas APOE2 homozygotes are protected from late-onset AD. It was hypothesized that expression of the "protective" APOE2 variant by genetic modification of the central nervous system (CNS) of APOE4 homozygotes could reverse or prevent progressive neurologic damage. To assess the CNS distribution and safety of APOE2 gene therapy for AD in a large-animal model, intraparenchymal, intracisternal, and intraventricular routes of delivery to the CNS of nonhuman primates of AAVrh.10hAPOE2-HA, an AAVrh.10 serotype coding for an HA-tagged human APOE2 cDNA sequence, were evaluated. To evaluate the route of delivery that achieves the widest extent of APOE2 expression in the CNS, the expression of APOE2 in the CNS was evaluated 2 months following vector administration for APOE2 DNA, mRNA, and protein. Finally, using conventional toxicology assays, the safety of the best route of delivery was assessed. The data demonstrated that while all three routes are capable of mediating ApoE2 expression in AD relevant regions, intracisternal delivery of AAVrh.10hAPOE2-HA safely mediated wide distribution of ApoE2 with the least invasive surgical intervention, thus providing the optimal strategy to deliver vector-mediated human APOE2 to the CNS.
Collapse
Affiliation(s)
- Jonathan B Rosenberg
- 1 Department of Genetic Medicine, Weill Cornell Medical College , New York, New York
| | - Michael G Kaplitt
- 2 Department of Neurosurgery, Weill Cornell Medical College , New York, New York
| | - Bishnu P De
- 1 Department of Genetic Medicine, Weill Cornell Medical College , New York, New York
| | - Alvin Chen
- 1 Department of Genetic Medicine, Weill Cornell Medical College , New York, New York
| | - Thomas Flagiello
- 1 Department of Genetic Medicine, Weill Cornell Medical College , New York, New York
| | - Christiana Salami
- 1 Department of Genetic Medicine, Weill Cornell Medical College , New York, New York
| | - Eduard Pey
- 1 Department of Genetic Medicine, Weill Cornell Medical College , New York, New York
| | - Lingzhi Zhao
- 3 Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medical College , New York, New York
| | - Rodolfo J Ricart Arbona
- Center of Comparative Medicine and Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sebastien Monette
- Laboratory of Comparative Pathology, Memorial Sloan Kettering Cancer Center, The Rockefeller University , Weill Cornell Medical College, New York, New York
| | - Jonathan P Dyke
- 6 Department of Radiology, Weill Cornell Medical College , New York, New York
| | - Douglas J Ballon
- 1 Department of Genetic Medicine, Weill Cornell Medical College , New York, New York.,6 Department of Radiology, Weill Cornell Medical College , New York, New York
| | - Stephen M Kaminsky
- 1 Department of Genetic Medicine, Weill Cornell Medical College , New York, New York
| | - Dolan Sondhi
- 1 Department of Genetic Medicine, Weill Cornell Medical College , New York, New York
| | - Gregory A Petsko
- 3 Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medical College , New York, New York
| | - Steven M Paul
- 7 Voyager Therapeutics, Inc. , Cambridge, Massachusetts
| | - Ronald G Crystal
- 1 Department of Genetic Medicine, Weill Cornell Medical College , New York, New York
| |
Collapse
|
74
|
Dietary betaine supplementation in hens modulates hypothalamic expression of cholesterol metabolic genes in F1 cockerels through modification of DNA methylation. Comp Biochem Physiol B Biochem Mol Biol 2018; 217:14-20. [DOI: 10.1016/j.cbpb.2017.12.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 11/28/2017] [Accepted: 12/05/2017] [Indexed: 11/20/2022]
|
75
|
Diotel N, Charlier TD, Lefebvre d'Hellencourt C, Couret D, Trudeau VL, Nicolau JC, Meilhac O, Kah O, Pellegrini E. Steroid Transport, Local Synthesis, and Signaling within the Brain: Roles in Neurogenesis, Neuroprotection, and Sexual Behaviors. Front Neurosci 2018; 12:84. [PMID: 29515356 PMCID: PMC5826223 DOI: 10.3389/fnins.2018.00084] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 02/02/2018] [Indexed: 01/18/2023] Open
Abstract
Sex steroid hormones are synthesized from cholesterol and exert pleiotropic effects notably in the central nervous system. Pioneering studies from Baulieu and colleagues have suggested that steroids are also locally-synthesized in the brain. Such steroids, called neurosteroids, can rapidly modulate neuronal excitability and functions, brain plasticity, and behavior. Accumulating data obtained on a wide variety of species demonstrate that neurosteroidogenesis is an evolutionary conserved feature across fish, birds, and mammals. In this review, we will first document neurosteroidogenesis and steroid signaling for estrogens, progestagens, and androgens in the brain of teleost fish, birds, and mammals. We will next consider the effects of sex steroids in homeostatic and regenerative neurogenesis, in neuroprotection, and in sexual behaviors. In a last part, we will discuss the transport of steroids and lipoproteins from the periphery within the brain (and vice-versa) and document their effects on the blood-brain barrier (BBB) permeability and on neuroprotection. We will emphasize the potential interaction between lipoproteins and sex steroids, addressing the beneficial effects of steroids and lipoproteins, particularly HDL-cholesterol, against the breakdown of the BBB reported to occur during brain ischemic stroke. We will consequently highlight the potential anti-inflammatory, anti-oxidant, and neuroprotective properties of sex steroid and lipoproteins, these latest improving cholesterol and steroid ester transport within the brain after insults.
Collapse
Affiliation(s)
- Nicolas Diotel
- Université de La Réunion, Institut National de la Santé et de la Recherche Médicale, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien, Saint-Denis de La Réunion, France
| | - Thierry D. Charlier
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes, France
| | - Christian Lefebvre d'Hellencourt
- Université de La Réunion, Institut National de la Santé et de la Recherche Médicale, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien, Saint-Denis de La Réunion, France
| | - David Couret
- Université de La Réunion, Institut National de la Santé et de la Recherche Médicale, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien, Saint-Denis de La Réunion, France
- CHU de La Réunion, Saint-Denis, France
| | | | - Joel C. Nicolau
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes, France
| | - Olivier Meilhac
- Université de La Réunion, Institut National de la Santé et de la Recherche Médicale, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien, Saint-Denis de La Réunion, France
- CHU de La Réunion, Saint-Denis, France
| | - Olivier Kah
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes, France
| | - Elisabeth Pellegrini
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes, France
| |
Collapse
|
76
|
Chen CPC, Huang YC, Chang CN, Chen JL, Hsu CC, Lin WY. Changes of cerebrospinal fluid protein concentrations and gait patterns in geriatric normal pressure hydrocephalus patients after ventriculoperitoneal shunting surgery. Exp Gerontol 2018; 106:109-115. [PMID: 29408782 DOI: 10.1016/j.exger.2018.01.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 01/07/2018] [Accepted: 01/31/2018] [Indexed: 10/18/2022]
Abstract
Normal pressure hydrocephalus (NPH) was the first type of dementia ever described that can be treated using ventriculoperitoneal shunting surgery. Three typical clinical symptoms of NPH include gait disturbance, progressive cognitive dysfunction, and urinary incontinence. Although there are articles that have discovered several cerebrospinal fluid (CSF) protein biomarkers associated with NPH; however, studies examining individual and total protein concentrations from the ventricular CSF before and after shunting surgery are lacking. This study used proteomics to calculate the CSF individual and total protein concentrations before, and one week, one month and three months after the shunting surgery. Parameters of cadence, step length, walking speed, and percentages of single- and double-limb support in a gait cycle were measured. Protein concentrations associated with anti-oxidation, aging, and in the prevention of neurotoxic agent production increased by at least 2-folds after the surgery, indicating that the brain may become less susceptible to neurodegeneration. These proteins were alpha-1B-glycoprotein, apolipoproteins A-1 & A-IV, prostaglandin-H2 D-isomerase, alpha-1-antitrypsin, and serotransferrin. In gait analysis, lower cadence, decreased double-limb support, longer step length, and increased single-limb support were observed after the surgery, indicating a more stable walking balance. These changes lasted for a period of at least 3 months. As a result, shunting surgery may be recommended for geriatric patients with confirmed diagnosis of normal pressure hydrocephalus.
Collapse
Affiliation(s)
- Carl P C Chen
- Department of Physical Medicine & Rehabilitation, Chang Gung Memorial Hospital at Linkou and College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan City, Taiwan.
| | - Yin-Cheng Huang
- Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou and College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan City, Taiwan
| | - Chen-Nen Chang
- Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou and College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan City, Taiwan
| | - Jean-Lon Chen
- Department of Physical Medicine & Rehabilitation, Chang Gung Memorial Hospital at Linkou and College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan City, Taiwan
| | - Chih-Chin Hsu
- Department of Physical Medicine & Rehabilitation, Chang Gung Memorial Hospital at Keelung and College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan City, Taiwan
| | - Wan-Ying Lin
- Department of Physical Medicine & Rehabilitation, Chang Gung Memorial Hospital at Linkou and College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan City, Taiwan
| |
Collapse
|
77
|
A Prospective Birth Cohort Study on Maternal Cholesterol Levels and Offspring Attention Deficit Hyperactivity Disorder: New Insight on Sex Differences. Brain Sci 2017; 8:brainsci8010003. [PMID: 29295472 PMCID: PMC5789334 DOI: 10.3390/brainsci8010003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 12/19/2017] [Accepted: 12/20/2017] [Indexed: 01/15/2023] Open
Abstract
Growing evidence suggests that maternal cholesterol levels are important in the offspring’s brain growth and development. Previous studies on cholesterols and brain functions were mostly in adults. We sought to examine the prospective association between maternal cholesterol levels and the risk of attention deficit hyperactivity disorder (ADHD) in the offspring. We analyzed data from the Boston Birth Cohort, enrolled at birth and followed from birth up to age 15 years. The final analyses included 1479 mother-infant pairs: 303 children with ADHD, and 1176 neurotypical children without clinician-diagnosed neurodevelopmental disorders. The median age of the first diagnosis of ADHD was seven years. The multiple logistic regression results showed that a low maternal high-density lipoprotein level (≤60 mg/dL) was associated with an increased risk of ADHD, compared to a higher maternal high-density lipoprotein level, after adjusting for pertinent covariables. A “J” shaped relationship was observed between triglycerides and ADHD risk. The associations with ADHD for maternal high-density lipoprotein and triglycerides were more pronounced among boys. The findings based on this predominantly urban low-income minority birth cohort raise a new mechanistic perspective for understanding the origins of ADHD and the gender differences and future targets in the prevention of ADHD.
Collapse
|
78
|
Fung KY, Wang C, Nyegaard S, Heit B, Fairn GD, Lee WL. SR-BI Mediated Transcytosis of HDL in Brain Microvascular Endothelial Cells Is Independent of Caveolin, Clathrin, and PDZK1. Front Physiol 2017; 8:841. [PMID: 29163190 PMCID: PMC5670330 DOI: 10.3389/fphys.2017.00841] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 10/09/2017] [Indexed: 01/19/2023] Open
Abstract
The vascular endothelium supplying the brain exhibits very low paracellular and transcellular permeability and is a major constituent of the blood-brain barrier. High-density lipoprotein (HDL) crosses the blood-brain barrier by transcytosis, but technical limitations have made it difficult to elucidate its regulation. Using a combination of spinning-disc confocal and total internal reflection fluorescence microscopy, we examined the uptake and transcytosis of HDL by human primary brain microvascular endothelial cell monolayers. Using these approaches, we report that HDL internalization requires dynamin but not clathrin heavy chain and that its internalization and transcytosis are saturable. Internalized HDL partially co-localized with the scavenger receptor BI (SR-BI) and knockdown of SR-BI significantly attenuated HDL internalization. However, we observed that the adaptor protein PDZK1—which is critical to HDL-SR-BI signaling in other tissues—is not required for HDL uptake in these cells. Additionally, while these cells express caveolin, the abundance of caveolae in this tissue is negligible and we find that SR-BI and caveolin do not co-fractionate. Furthermore, direct silencing of caveolin-1 had no impact on the uptake of HDL. Finally, inhibition of endothelial nitric oxide synthase increased HDL internalization while increasing nitric oxide levels had no impact. Together, these data indicate that SR-BI-mediated transcytosis in brain microvascular endothelial cells is distinct from uptake and signaling pathways described for this receptor in other cell types.
Collapse
Affiliation(s)
- Karen Y Fung
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada.,Keenan Research Center for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada
| | - Changsen Wang
- Keenan Research Center for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada
| | - Steffen Nyegaard
- Program in Cell Biology, Hospital for Sick Children, Toronto, ON, Canada
| | - Bryan Heit
- Department of Microbiology and Immunology, Centre for Human Immunology, University of Western Ontario, London, ON, Canada
| | - Gregory D Fairn
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada.,Keenan Research Center for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada.,Department of Surgery, University of Toronto, ON, Canada
| | - Warren L Lee
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada.,Keenan Research Center for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada.,Departments of Medicine and Laboratory Medicine and Pathobiology, University of Toronto, ON, Canada
| |
Collapse
|
79
|
Tai LM, Balu D, Avila-Munoz E, Abdullah L, Thomas R, Collins N, Valencia-Olvera AC, LaDu MJ. EFAD transgenic mice as a human APOE relevant preclinical model of Alzheimer's disease. J Lipid Res 2017; 58:1733-1755. [PMID: 28389477 PMCID: PMC5580905 DOI: 10.1194/jlr.r076315] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 04/06/2017] [Indexed: 01/12/2023] Open
Abstract
Identified in 1993, APOE4 is the greatest genetic risk factor for sporadic Alzheimer's disease (AD), increasing risk up to 15-fold compared with APOE3, with APOE2 decreasing AD risk. However, the functional effects of APOE4 on AD pathology remain unclear and, in some cases, controversial. In vivo progress to understand how the human (h)-APOE genotypes affect AD pathology has been limited by the lack of a tractable familial AD-transgenic (FAD-Tg) mouse model expressing h-APOE rather than mouse (m)-APOE. The disparity between m- and h-apoE is relevant for virtually every AD-relevant pathway, including amyloid-β (Aβ) deposition and clearance, neuroinflammation, tau pathology, neural plasticity and cerebrovascular deficits. EFAD mice were designed as a temporally useful preclinical FAD-Tg-mouse model expressing the h-APOE genotypes for identifying mechanisms underlying APOE-modulated symptoms of AD pathology. From their first description in 2012, EFAD mice have enabled critical basic and therapeutic research. Here we review insights gleaned from the EFAD mice and summarize future directions.
Collapse
Affiliation(s)
- Leon M Tai
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612
| | - Deebika Balu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612
| | - Evangelina Avila-Munoz
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612
| | | | - Riya Thomas
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612
| | - Nicole Collins
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612
| | | | - Mary Jo LaDu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612.
| |
Collapse
|
80
|
Koch M, Furtado JD, Falk K, Leypoldt F, Mukamal KJ, Jensen MK. Apolipoproteins and their subspecies in human cerebrospinal fluid and plasma. ALZHEIMER'S & DEMENTIA: DIAGNOSIS, ASSESSMENT & DISEASE MONITORING 2017; 6:182-187. [PMID: 28289700 PMCID: PMC5338868 DOI: 10.1016/j.dadm.2017.01.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
INTRODUCTION Subspecies of apolipoproteins can be defined by fractionating apolipoproteins based on the presence and absence of coexisting apolipoproteins. METHODS We determined age- and sex-adjusted correlations of enzyme-linked immunosorbent assay-measured plasma and cerebrospinal fluid (CSF) apolipoproteins (apoA-I, apoC-III, apoE, and apoJ) or apolipoprotein subspecies (apoA-I with and without apoC-III, ApoE, or apoJ; apoE with and without apoC-III or apoJ) in 22 dementia-free participants. RESULTS CSF apoE did not correlate with plasma apolipoproteins or their subspecies. CSF apoJ correlated most strongly with plasma apoA-I without apoJ (r = 0.7). CSF apoA-I correlated similarly strong with plasma total apoA-I and all apoA-I subspecies (r ≥ 0.4) except for apoA-I with apoE (r = 0.3) or apoA-I with apoJ (r = 0.3). CSF apoC-III was most strongly correlated with plasma apoA-I with apoC-III (r = 0.7). DISCUSSION CSF levels of some apolipoproteins implicated in the pathophysiology of dementia might be better approximated by specific plasma apolipoprotein subspecies than total plasma concentrations.
Collapse
Affiliation(s)
- Manja Koch
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Jeremy D Furtado
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Kim Falk
- Neuroimmunology Unit, Christian-Albrechts-University Kiel, Kiel, Germany; Department of Neurology, Institute of Clinical Chemistry, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Frank Leypoldt
- Neuroimmunology Unit, Christian-Albrechts-University Kiel, Kiel, Germany; Department of Neurology, Institute of Clinical Chemistry, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Kenneth J Mukamal
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Majken K Jensen
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Medicine, Channing Division of Network Medicine, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| |
Collapse
|
81
|
Weiler A, Volkenhoff A, Hertenstein H, Schirmeier S. Metabolite transport across the mammalian and insect brain diffusion barriers. Neurobiol Dis 2017; 107:15-31. [PMID: 28237316 DOI: 10.1016/j.nbd.2017.02.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 01/02/2017] [Accepted: 02/20/2017] [Indexed: 12/31/2022] Open
Abstract
The nervous system in higher vertebrates is separated from the circulation by a layer of specialized endothelial cells. It protects the sensitive neurons from harmful blood-derived substances, high and fluctuating ion concentrations, xenobiotics or even pathogens. To this end, the brain endothelial cells and their interlinking tight junctions build an efficient diffusion barrier. A structurally analogous diffusion barrier exists in insects, where glial cell layers separate the hemolymph from the neural cells. Both types of diffusion barriers, of course, also prevent influx of metabolites from the circulation. Because neuronal function consumes vast amounts of energy and necessitates influx of diverse substrates and metabolites, tightly regulated transport systems must ensure a constant metabolite supply. Here, we review the current knowledge about transport systems that carry key metabolites, amino acids, lipids and carbohydrates into the vertebrate and Drosophila brain and how this transport is regulated. Blood-brain and hemolymph-brain transport functions are conserved and we can thus use a simple, genetically accessible model system to learn more about features and dynamics of metabolite transport into the brain.
Collapse
Affiliation(s)
- Astrid Weiler
- Institut für Neuro- und Verhaltensbiologie, Universität Münster, Badestr. 9, 48149 Münster, Germany
| | - Anne Volkenhoff
- Institut für Neuro- und Verhaltensbiologie, Universität Münster, Badestr. 9, 48149 Münster, Germany
| | - Helen Hertenstein
- Institut für Neuro- und Verhaltensbiologie, Universität Münster, Badestr. 9, 48149 Münster, Germany
| | - Stefanie Schirmeier
- Institut für Neuro- und Verhaltensbiologie, Universität Münster, Badestr. 9, 48149 Münster, Germany.
| |
Collapse
|
82
|
Slot RE, Van Harten AC, Kester MI, Jongbloed W, Bouwman FH, Teunissen CE, Scheltens P, Veerhuis R, van der Flier WM. Apolipoprotein A1 in Cerebrospinal Fluid and Plasma and Progression to Alzheimer’s Disease in Non-Demented Elderly. J Alzheimers Dis 2017; 56:687-697. [DOI: 10.3233/jad-151068] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Rosalinde E.R. Slot
- Department of Neurology and Alzheimer Center, VU University Medical Center, Amsterdam, The Netherlands
| | - Argonde C. Van Harten
- Department of Neurology and Alzheimer Center, VU University Medical Center, Amsterdam, The Netherlands
| | - Maartje I. Kester
- Department of Neurology and Alzheimer Center, VU University Medical Center, Amsterdam, The Netherlands
| | - Wesley Jongbloed
- Department of Clinical Chemistry, Neurochemistry Laboratory, VU University Medical Center, Amsterdam, The Netherlands
| | - Femke H. Bouwman
- Department of Neurology and Alzheimer Center, VU University Medical Center, Amsterdam, The Netherlands
| | - Charlotte E. Teunissen
- Department of Clinical Chemistry, Neurochemistry Laboratory, VU University Medical Center, Amsterdam, The Netherlands
| | - Philip Scheltens
- Department of Neurology and Alzheimer Center, VU University Medical Center, Amsterdam, The Netherlands
| | - Robert Veerhuis
- Department of Clinical Chemistry, Neurochemistry Laboratory, VU University Medical Center, Amsterdam, The Netherlands
- Department of Psychiatry, VU University Medical Center, Amsterdam, The Netherlands
| | - Wiesje M. van der Flier
- Department of Neurology and Alzheimer Center, VU University Medical Center, Amsterdam, The Netherlands
- Department of Epidemiology and Biostatistics, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
83
|
Heinsinger NM, Gachechiladze MA, Rebeck GW. Apolipoprotein E Genotype Affects Size of ApoE Complexes in Cerebrospinal Fluid. J Neuropathol Exp Neurol 2016; 75:918-924. [PMID: 27516118 DOI: 10.1093/jnen/nlw067] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Apolipoprotein E (apoE) is associated with lipoproteins in the cerebrospinal fluid (CSF). APOE4 increases and APOE2 decreases the risk for Alzheimer disease (AD) compared to the risk associated with APOE3 Because apoE4 is less efficient at cholesterol efflux than apoE2 or apoE3 in vitro, we hypothesized that APOE genotype may affect apoE particle size in vivo and that these size differences may be related to AD risk. We used nondenaturing gel electrophoresis to test for differences in the size of apoE complexes in human CSF samples of various APOE genotypes and created profiles of each sample to compare the patterns of apoE distribution. For middle-aged adults with no dementia, APOE 2.3 individuals had significantly larger apoE complexes than APOE 3.3 subjects, who had significantly larger apoE complexes than APOE 3.4 and APOE 4.4 individuals. Similarly, in an independent cohort of older adults, CSF apoE complexes of APOE4-positive individuals were smaller than those of the APOE4-negative individuals. Compared to individuals with no dementia, those with the mildest stages of dementia had similar sized CSF apoE complexes. These results identify a novel phenotypic difference in the size of CSF apoE complexes in middle age that correlate with the risk of AD later in life.
Collapse
Affiliation(s)
- Nicolette Mary Heinsinger
- From the Department of Biology (NMH, MAG); and Department of Neuroscience, Georgetown University (GWR), Washington, District of Columbia
| | - Mariam Alexandra Gachechiladze
- From the Department of Biology (NMH, MAG); and Department of Neuroscience, Georgetown University (GWR), Washington, District of Columbia
| | - G William Rebeck
- From the Department of Biology (NMH, MAG); and Department of Neuroscience, Georgetown University (GWR), Washington, District of Columbia
| |
Collapse
|
84
|
Ferretti G, Bacchetti T, Principi F, Di Ludovico F, Viti B, Angeleri VA, Danni M, Provinciali L. Increased levels of lipid hydroperoxides in plasma of patients with multiple sclerosis: a relationship with paraoxonase activity. Mult Scler 2016; 11:677-82. [PMID: 16320727 DOI: 10.1191/1352458505ms1240oa] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Paraoxonase, an enzyme associated with high density lipoproteins (HDL), plays an important role in the anti-oxidant and anti-inflammatory properties exerted by HDL. Increasing evidence supports a role of free radicals and oxidative stress in the inflammatory processes and in the pathogenesis of multiple sclerosis (MS). The aim of this study was to further investigate the relationship between oxidative damage and MS; therefore we compared the paraoxonase activity and levels of cholesteryl ester hydroperoxides (CE-OOH), as marker of lipid peroxidation, in plasma isolated from healthy subjects (n=89) and from MS patients (n=24) in the early stage disability (EDSSB<3.5). Our results demonstrated for the first time that the activity of paraoxonase in the plasma of MS subjects was significantly lower with respect to controls (p<0.001). Moreover, our results showed a significant increase in the levels of CE-OOH in plasma from MS subjects (p<0.001). CE-OOH are biologically active substances derived from the oxidation of cholesteryl ester localized in the hydrophobic core of plasma lipoproteins (HDL, LDL). Therefore, our study demonstrates alterations of lipoprotein peroxidation in MS and provides further evidence that oxidative stress and impairment of the anti-oxidant system may play a role in MS.
Collapse
Affiliation(s)
- G Ferretti
- Facoltà di Medicina e Chirurgia, Istituto di Biochimica, Università Politecnica delle Marche, Ancona, Italia.
| | | | | | | | | | | | | | | |
Collapse
|
85
|
Najyb O, Do Carmo S, Alikashani A, Rassart E. Apolipoprotein D Overexpression Protects Against Kainate-Induced Neurotoxicity in Mice. Mol Neurobiol 2016; 54:3948-3963. [PMID: 27271124 PMCID: PMC7091089 DOI: 10.1007/s12035-016-9920-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Accepted: 05/03/2016] [Indexed: 01/23/2023]
Abstract
Excitotoxicity due to the excessive activation of glutamatergic receptors leads to neuronal dysfunction and death. Excitotoxicity has been implicated in the pathogenesis of a myriad of neurodegenerative diseases with distinct etiologies such as Alzheimer's and Parkinson's. Numerous studies link apolipoprotein D (apoD), a secreted glycoprotein highly expressed in the central nervous system (CNS), to maintain and protect neurons in various mouse models of acute stress and neurodegeneration. Here, we used a mouse model overexpressing human apoD in neurons (H-apoD Tg) to test the neuroprotective effects of apoD in the kainic acid (KA)-lesioned hippocampus. Our results show that apoD overexpression in H-apoD Tg mice induces an increased resistance to KA-induced seizures, significantly attenuates inflammatory responses and confers protection against KA-induced cell apoptosis in the hippocampus. The apoD-mediated protection against KA-induced toxicity is imputable in part to increased plasma membrane Ca2+ ATPase type 2 expression (1.7-fold), decreased N-methyl-D-aspartate receptor (NMDAR) subunit NR2B levels (30 %) and lipid metabolism alterations. Indeed, we demonstrate that apoD can attenuate intracellular cholesterol content in primary hippocampal neurons and in brain of H-apoD Tg mice. In addition, apoD can be internalised by neurons and this internalisation is accentuated in ageing and injury conditions. Our results provide additional mechanistic information on the apoD-mediated neuroprotection in neurodegenerative conditions.
Collapse
Affiliation(s)
- Ouafa Najyb
- Laboratoire de Biologie Moléculaire, Département des Sciences Biologiques, Centre BioMed, Université du Québec à Montréal, Case Postale 8888, Succursale Centre-ville, Montréal, QC, H3C-3P8, Canada
| | - Sonia Do Carmo
- Laboratoire de Biologie Moléculaire, Département des Sciences Biologiques, Centre BioMed, Université du Québec à Montréal, Case Postale 8888, Succursale Centre-ville, Montréal, QC, H3C-3P8, Canada
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Azadeh Alikashani
- Laboratoire de Biologie Moléculaire, Département des Sciences Biologiques, Centre BioMed, Université du Québec à Montréal, Case Postale 8888, Succursale Centre-ville, Montréal, QC, H3C-3P8, Canada
| | - Eric Rassart
- Laboratoire de Biologie Moléculaire, Département des Sciences Biologiques, Centre BioMed, Université du Québec à Montréal, Case Postale 8888, Succursale Centre-ville, Montréal, QC, H3C-3P8, Canada.
| |
Collapse
|
86
|
Abstract
PURPOSE OF REVIEW This article evaluates recent experimental and human evidence regarding the involvement of lipids, lipoproteins, and apolipoproteins in neurodegenerative diseases, and reviews the current literature of the effects of cholesterol-lowering treatment on cognition. RECENT FINDINGS Plasma levels of traditional lipids and lipoproteins are not consistently associated with risk of dementia even though low plasma levels of apolipoprotein E, through unknown mechanisms, robustly predict future dementia. Experimental evidence suggests neuroprotective roles of several brain and cerebrospinal fluid apolipoproteins. Whether plasma levels of apolipoprotein E, or any other apolipoprotein with possible central nervous system and/or blood-brain barrier functions (apolipoproteins J, A-I, A-II, A-IV, D, C-I, and C-III) may become accessible biomarker components that improve risk prediction for dementia together with genetic risk variants and cardiovascular risk factors remains to be determined. SUMMARY Apolipoproteins with well established functions in peripheral lipid metabolism may play important roles for brain vascular health and Alzheimer's disease pathophysiology. Experimental work on lipids, lipoproteins, and apolipoproteins in the central nervous system together with robust prospective human studies will help to substantiate the drug target potential of these lipid components.
Collapse
Affiliation(s)
- Cheryl L Wellington
- aDepartment of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada bDepartment of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospitals cFaculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | |
Collapse
|
87
|
Mahley RW. Central Nervous System Lipoproteins: ApoE and Regulation of Cholesterol Metabolism. Arterioscler Thromb Vasc Biol 2016; 36:1305-15. [PMID: 27174096 DOI: 10.1161/atvbaha.116.307023] [Citation(s) in RCA: 314] [Impact Index Per Article: 34.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Accepted: 04/29/2016] [Indexed: 12/19/2022]
Abstract
ApoE on high-density lipoproteins is primarily responsible for lipid transport and cholesterol homeostasis in the central nervous system (CNS). Normally produced mostly by astrocytes, apoE is also produced under neuropathologic conditions by neurons. ApoE on high-density lipoproteins is critical in redistributing cholesterol and phospholipids for membrane repair and remodeling. The 3 main structural isoforms differ in their effectiveness. Unlike apoE2 and apoE3, apoE4 has markedly altered CNS metabolism, is associated with Alzheimer disease and other neurodegenerative disorders, and is expressed at lower levels in brain and cerebrospinal fluid. ApoE4-expressing cultured astrocytes and neurons have reduced cholesterol and phospholipid secretion, decreased lipid-binding capacity, and increased intracellular degradation. Two structural features are responsible for apoE4 dysfunction: domain interaction, in which arginine-61 interacts ionically with glutamic acid-255, and a less stable conformation than apoE3 and apoE2. Blocking domain interaction by gene targeting (replacing arginine-61 with threonine) or by small-molecule structure correctors increases CNS apoE4 levels and lipid-binding capacity and decreases intracellular degradation. Small molecules (drugs) that disrupt domain interaction, so-called structure correctors, could prevent the apoE4-associated neuropathology by blocking the formation of neurotoxic fragments. Understanding how to modulate CNS cholesterol transport and metabolism is providing important insights into CNS health and disease.
Collapse
Affiliation(s)
- Robert W Mahley
- From the Gladstone Institute of Neurological Disease, San Francisco, CA; and Departments of Pathology and Medicine, University of California, San Francisco.
| |
Collapse
|
88
|
Yoon H, Flores LF, Kim J. MicroRNAs in brain cholesterol metabolism and their implications for Alzheimer's disease. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:2139-2147. [PMID: 27155217 DOI: 10.1016/j.bbalip.2016.04.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 04/28/2016] [Accepted: 04/29/2016] [Indexed: 01/01/2023]
Abstract
Cholesterol is important for various neuronal functions in the brain. Brain has elaborate regulatory mechanisms to control cholesterol metabolism that are distinct from the mechanisms in periphery. Interestingly, dysregulation of the cholesterol metabolism is strongly associated with a number of neurodegenerative diseases. MicroRNAs are short non-coding RNAs acting as post-transcriptional gene regulators. Recently, several microRNAs are demonstrated to be involved in regulating cholesterol metabolism in the brain. This article reviews the regulatory mechanisms of cellular cholesterol homeostasis in the brain. In addition, we discuss the role of microRNAs in brain cholesterol metabolism and their potential implications for the treatment of Alzheimer's disease. This article is part of a special issue entitled: MicroRNAs and lipid/energy metabolism and related diseases edited by Carlos Fernández-Hernando and Yajaira Suárez.
Collapse
Affiliation(s)
- Hyejin Yoon
- Neurobiology of Disease Graduate Program, Mayo Graduate School, Jacksonville, FL, United States; Department of Neuroscience, Mayo Clinic, Jacksonville, FL, United States
| | - Luis F Flores
- Biochemistry and Molecular Biology Graduate Program, Mayo Graduate School, Jacksonville, FL, United States
| | - Jungsu Kim
- Neurobiology of Disease Graduate Program, Mayo Graduate School, Jacksonville, FL, United States; Department of Neuroscience, Mayo Clinic, Jacksonville, FL, United States.
| |
Collapse
|
89
|
Berland C, Cansell C, Hnasko TS, Magnan C, Luquet S. Dietary triglycerides as signaling molecules that influence reward and motivation. Curr Opin Behav Sci 2016; 9:126-135. [PMID: 28191490 DOI: 10.1016/j.cobeha.2016.03.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The reinforcing and motivational aspects of food are tied to the release of the dopamine in the mesolimbic system (ML). Free fatty acids from triglyceride (TG)-rich particles are released upon action of TG-lipases found at high levels in peripheral oxidative tissue (muscle, heart), but also in the ML. This suggests that local TG-hydrolysis in the ML might regulate food seeking and reward. Indeed, evidence now suggests that dietary TG directly target the ML to regulate amphetamine-induced locomotion and reward seeking behavior. Though the cellular mechanisms of TG action are unresolved, TG act in part through ML lipoprotein lipase, upstream of dopamine 2 receptor (D2R), and show desensitization in conditions of chronically elevated plasma TG as occur in obesity. TG sensing in the ML therefore represents a new mechanism by which chronic consumption of dietary fat might lead to adaptations in the ML and dysregulated feeding behaviors.
Collapse
Affiliation(s)
- Chloé Berland
- Univ Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, F-75205 Paris, France; Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health, München/Neuherberg, Germany; Div. of Metabolic Diseases, Dept. of Medicine, Technische Universität München, Germany
| | - Céline Cansell
- Université de Nice Sophia Antipolis, IPMC, Sophia Antipolis, F-06560, France; CNRS, IPMC, Sophia Antipolis, F-06560, France
| | - Thomas S Hnasko
- Department of Neurosciences, University of California, San Diego, La Jolla CA, USA
| | - Christophe Magnan
- Univ Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, F-75205 Paris, France
| | - Serge Luquet
- Univ Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, F-75205 Paris, France
| |
Collapse
|
90
|
Yassine HN, Feng Q, Chiang J, Petrosspour LM, Fonteh AN, Chui HC, Harrington MG. ABCA1-Mediated Cholesterol Efflux Capacity to Cerebrospinal Fluid Is Reduced in Patients With Mild Cognitive Impairment and Alzheimer's Disease. J Am Heart Assoc 2016; 5:JAHA.115.002886. [PMID: 26873692 PMCID: PMC4802440 DOI: 10.1161/jaha.115.002886] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Background Animal and human studies indicate that ABCA1‐mediated cholesterol transport is important in Alzheimer's disease (AD). We hypothesized that the efficiency of cerebrospinal fluid (CSF) to facilitate ABCA1‐mediated cholesterol efflux would be reduced in participants with mild cognitive impairment (MCI) or AD compared with cognitively healthy participants. Methods and Results CSF was collected from a cross‐sectional study of cognitively healthy participants (n=47) and participants with MCI (n=35) or probable AD (n=26).The capacity of CSF to mediate cholesterol transport was assessed using a BHK cell line that can be induced to express the ABCA1 transporter. ABCA1‐mediated cholesterol efflux capacity was 30% less in participants with MCI or AD compared with cognitively healthy participants (P<0.001 for both). Cholesterol efflux capacity correlated with CSF cholesterol content (r=0.37, P<0.001). CSF phosphatidylcholine decreased in participants with MCI and AD compared with cognitively healthy participants (9% less in MCI and 27% less in AD compared with cognitively healthy participants, P=0.01) and correlated with CSF efflux capacity (r=0.3, P=0.001). CSF sphingomyelin also correlated with the efflux capacity (r=0.24, P=0.02). Concentrations of CSF apoA‐I and apoE did not significantly correlate with measures of efflux capacity. Conclusions In people with MCI and AD, the capacity of CSF to facilitate ABCA1‐mediated cholesterol efflux is impaired. This lesser cholesterol efflux in MCI supports a pathophysiological role for ABCA1‐mediated cholesterol transport in early neurodegeneration.
Collapse
Affiliation(s)
- Hussein N Yassine
- Department of Medicine, University of Southern California, Los Angeles, CA
| | - Qingru Feng
- Department of Medicine, University of Southern California, Los Angeles, CA
| | - Jiarong Chiang
- Molecular Neurology Program, Huntington Medical Research Institutes, Pasadena, CA
| | - Larissa M Petrosspour
- Department of Medicine, University of Southern California, Los Angeles, CA Department of Neurology, University of Southern California, Los Angeles, CA
| | - Alfred N Fonteh
- Molecular Neurology Program, Huntington Medical Research Institutes, Pasadena, CA
| | - Helena C Chui
- Department of Neurology, University of Southern California, Los Angeles, CA
| | - Michael G Harrington
- Molecular Neurology Program, Huntington Medical Research Institutes, Pasadena, CA
| |
Collapse
|
91
|
Abstract
PURPOSE OF REVIEW Dementia is a major cause of disability and institutionalization. Apart from age and apolipoprotein E (APOE) genotype, there are currently no established, clinically relevant, noninvasive markers of dementia. We conducted a literature search of recent observational epidemiological studies evaluating the relevance of HDL cholesterol (HDL-C) and apolipoproteins as biomarkers of future and prevalent risk of dementia. RECENT FINDINGS HDL-C and apolipoproteins, such as apoE have been suggested to play important roles in brain function and have been associated with dementia and Alzheimer's disease in observational studies. However, findings have been inconsistent, especially across study designs. In recent years, modern proteomic approaches have enabled the investigation of further apolipoproteins involved in the deposition and clearance of β-amyloid, a determining factor for subsequent neurodegeneration. SUMMARY Associations in cross-sectional studies are not always indicative of a prospective relationship. Large studies find that plasma HDL-C and apoE are inversely associated with dementia. Higher apoJ levels might be a marker of prevalent dementia, but were not associated with risk of future dementia. The investigation of HDL-C and apolipoproteins in relation to dementia represents an area of opportunity. Additional prospective studies that account for potential confounding factors and that explore potential effect modifiers such as APOE genotype and sex are needed to fully investigate the potential of these noninvasive measures in disease prediction.
Collapse
Affiliation(s)
- Manja Koch
- Harvard T.H. Chan School of Public Health, Department of Nutrition, Boston, Massachusetts, USA
| | - Majken K. Jensen
- Harvard T.H. Chan School of Public Health, Department of Nutrition, Boston, Massachusetts, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
92
|
Constantinou C, Karavia EA, Xepapadaki E, Petropoulou PI, Papakosta E, Karavyraki M, Zvintzou E, Theodoropoulos V, Filou S, Hatziri A, Kalogeropoulou C, Panayiotakopoulos G, Kypreos KE. Advances in high-density lipoprotein physiology: surprises, overturns, and promises. Am J Physiol Endocrinol Metab 2016; 310:E1-E14. [PMID: 26530157 DOI: 10.1152/ajpendo.00429.2015] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 10/30/2015] [Indexed: 12/21/2022]
Abstract
Emerging evidence strongly supports that changes in the HDL metabolic pathway, which result in changes in HDL proteome and function, appear to have a causative impact on a number of metabolic disorders. Here, we provide a critical review of the most recent and novel findings correlating HDL properties and functionality with various pathophysiological processes and disease states, such as obesity, type 2 diabetes mellitus, nonalcoholic fatty liver disease, inflammation and sepsis, bone and obstructive pulmonary diseases, and brain disorders.
Collapse
Affiliation(s)
| | - Eleni A Karavia
- Pharmacology Department, University of Patras Medical School, Rio Achaias, Greece
| | - Eva Xepapadaki
- Pharmacology Department, University of Patras Medical School, Rio Achaias, Greece
| | | | - Eugenia Papakosta
- Pharmacology Department, University of Patras Medical School, Rio Achaias, Greece
| | - Marilena Karavyraki
- Pharmacology Department, University of Patras Medical School, Rio Achaias, Greece
| | - Evangelia Zvintzou
- Pharmacology Department, University of Patras Medical School, Rio Achaias, Greece
| | | | - Serafoula Filou
- Pharmacology Department, University of Patras Medical School, Rio Achaias, Greece
| | - Aikaterini Hatziri
- Pharmacology Department, University of Patras Medical School, Rio Achaias, Greece
| | | | | | - Kyriakos E Kypreos
- Pharmacology Department, University of Patras Medical School, Rio Achaias, Greece
| |
Collapse
|
93
|
Siebel AL, Heywood SE, Kingwell BA. HDL and glucose metabolism: current evidence and therapeutic potential. Front Pharmacol 2015; 6:258. [PMID: 26582989 PMCID: PMC4628107 DOI: 10.3389/fphar.2015.00258] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 10/19/2015] [Indexed: 12/23/2022] Open
Abstract
High-density lipoprotein (HDL) and its principal apolipoprotein A-I (ApoA-I) have now been convincingly shown to influence glucose metabolism through multiple mechanisms. The key clinically relevant observations are that both acute HDL elevation via short-term reconstituted HDL (rHDL) infusion and chronically raising HDL via a cholesteryl ester transfer protein (CETP) inhibitor reduce blood glucose in individuals with type 2 diabetes mellitus (T2DM). HDL may mediate effects on glucose metabolism through actions in multiple organs (e.g., pancreas, skeletal muscle, heart, adipose, liver, brain) by three distinct mechanisms: (i) Insulin secretion from pancreatic beta cells, (ii) Insulin-independent glucose uptake, (iii) Insulin sensitivity. The molecular mechanisms appear to involve both direct HDL signaling actions as well as effects secondary to lipid removal from cells. The implications of glucoregulatory mechanisms linked to HDL extend from glycemic control to potential anti-ischemic actions via increased tissue glucose uptake and utilization. Such effects not only have implications for the prevention and management of diabetes, but also for ischemic vascular diseases including angina pectoris, intermittent claudication, cerebral ischemia and even some forms of dementia. This review will discuss the growing evidence for a role of HDL in glucose metabolism and outline related potential for HDL therapies.
Collapse
Affiliation(s)
- Andrew L Siebel
- Metabolic and Vascular Physiology Laboratory, Baker IDI Heart and Diabetes Institute , Melbourne, VIC, Australia
| | - Sarah Elizabeth Heywood
- Metabolic and Vascular Physiology Laboratory, Baker IDI Heart and Diabetes Institute , Melbourne, VIC, Australia
| | - Bronwyn A Kingwell
- Metabolic and Vascular Physiology Laboratory, Baker IDI Heart and Diabetes Institute , Melbourne, VIC, Australia
| |
Collapse
|
94
|
Robert J, Stukas S, Button E, Cheng WH, Lee M, Fan J, Wilkinson A, Kulic I, Wright SD, Wellington CL. Reconstituted high-density lipoproteins acutely reduce soluble brain Aβ levels in symptomatic APP/PS1 mice. Biochim Biophys Acta Mol Basis Dis 2015; 1862:1027-36. [PMID: 26454209 DOI: 10.1016/j.bbadis.2015.10.005] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 09/21/2015] [Accepted: 10/03/2015] [Indexed: 01/13/2023]
Abstract
Many lines of evidence suggest a protective role for high-density lipoprotein (HDL) and its major apolipoprotein (apo)A-I in Alzheimer's Disease (AD). HDL/apoA-I particles are produced by the liver and intestine and, in addition to removing excess cholesterol from the body, are increasingly recognized to have vasoprotective functions. Here we tested the ability of reconstituted HDL (rHDL) consisting of human apoA-I reconstituted with soy phosphatidylcholine for its ability to lower amyloid beta (Aβ) levels in symptomatic APP/PS1 mice, a well-characterized preclinical model of amyloidosis. Animals were treated intravenously either with four weekly doses (chronic study) or a single dose of 60mg/kg of rHDL (acute study). The major finding of our acute study is that soluble brain Aβ40 and Aβ42 levels were significantly reduced within 24h of a single dose of rHDL. By contrast, no changes were observed in our chronic study with respect to soluble or deposited Aβ levels in animals assessed 7days after the final weekly dose of rHDL, suggesting that beneficial effects diminish as rHDL is cleared from the body. Further, rHDL-treated animals showed no change in amyloid burden, cerebrospinal fluid (CSF) Aβ levels, neuroinflammation, or endothelial activation in the chronic study, suggesting that the pathology-modifying effects of rHDL may indeed be acute and may be specific to the soluble Aβ pool. That systemic administration of rHDL can acutely modify brain Aβ levels provides support for further investigation of the therapeutic potential of apoA-I-based agents for AD. This article is part of a Special Issue entitled: Vascular Contributions to Cognitive Impairment and Dementia edited by M. Paul Murphy, Roderick A. Corriveau and Donna M. Wilcock.
Collapse
Affiliation(s)
- Jérôme Robert
- Department of Pathology and Laboratory Medicine, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Sophie Stukas
- Department of Pathology and Laboratory Medicine, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Emily Button
- Department of Pathology and Laboratory Medicine, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Wai Hang Cheng
- Department of Pathology and Laboratory Medicine, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Michael Lee
- Department of Pathology and Laboratory Medicine, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jianjia Fan
- Department of Pathology and Laboratory Medicine, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Anna Wilkinson
- Department of Pathology and Laboratory Medicine, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Iva Kulic
- Department of Pathology and Laboratory Medicine, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Samuel D Wright
- Cardiovascular Therapeutics, CSL Limited, Parkville, Australia
| | - Cheryl L Wellington
- Department of Pathology and Laboratory Medicine, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
95
|
Abstract
Human cerebrospinal fluid (CSF) contains diverse lipid particles, including lipoproteins that are distinct from their plasma counterparts and contain apolipoprotein (apo) E isoforms, apoJ, and apoAI, and extracellular vesicles, which can be detected by annexin V binding. The aim of this study was to develop a method to quantify CSF particles and evaluate their relationship to aging and neurodegenerative diseases. We used a flow cytometric assay to detect annexin V-, apoE-, apoAI-, apoJ-, and amyloid (A) β42-positive particles in CSF from 131 research volunteers who were neurologically normal or had mild cognitive impairment (MCI), Alzheimer disease (AD) dementia, or Parkinson disease. APOE ε4/ε4 participants had CSF apoE-positive particles that were more frequently larger but at an 88% lower level versus those in APOE ε3/ε3 or APOE ε3/ε4 patients; this finding was reproduced in conditioned medium from mouse primary glial cell cultures with targeted replacement of apoE. Cerebrospinal fluid apoE-positive and β-amyloid (Aβ42)-positive particle concentrations were persistently reduced one-third to one-half in middle and older age subjects; apoAI-positive particle concentration progressively increased approximately 2-fold with age. Both apoAI-positive and annexin V-positive CSF particle levels were reduced one-third to one-half in CSF of MCI and/or AD dementia patients versus age-matched controls. Our approach provides new methods to investigate CNS lipid biology in relation to neurodegeneration and perhaps develop new biomarkers for diagnosis or treatment monitoring.
Collapse
|
96
|
Nakato M, Matsuo M, Kono N, Arita M, Arai H, Ogawa J, Kioka N, Ueda K. Neurite outgrowth stimulation by n-3 and n-6 PUFAs of phospholipids in apoE-containing lipoproteins secreted from glial cells. J Lipid Res 2015; 56:1880-90. [PMID: 26239183 DOI: 10.1194/jlr.m058164] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Indexed: 01/06/2023] Open
Abstract
PUFAs, which account for 25-30% of the total fatty acids in the human brain, are important for normal brain development and cognitive function. However, it remains unclear how PUFAs are delivered to neurons and exert their effects. In this study, we demonstrated that n-3 and n-6 PUFAs added to the medium are incorporated into membrane phospholipids of primary glial cells from rat cortices, and then secreted as the fatty acid moiety of phospholipids in apoE-containing lipoproteins (LpEs). Tandem mass spectrometry analysis further showed that LpEs secreted from glial cells contain a variety of metabolites of PUFAs produced in glial cells by elongation and unsaturation. LpEs are absorbed by endocytosis into neurons via LDL receptor-related protein 1. LpE-containing n-3 and n-6 PUFAs exhibit a strong effect on neurite outgrowth of hippocampal neurons by increasing the number of branches. This study sheds light on the novel role of LpEs in the central nervous system and also a novel pathway in which PUFAs act on neurons.
Collapse
Affiliation(s)
- Mitsuhiro Nakato
- Division of Applied Life Sciences, Kyoto University Graduate School of Agriculture, Kyoto 606-8502, Japan
| | - Michinori Matsuo
- Department of Food and Nutrition, Faculty of Home Economics, Kyoto Women's University, Kyoto 605-8501, Japan
| | - Nozomu Kono
- Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo 113-0033, Japan
| | - Makoto Arita
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Kanagawa 230-0045, Japan
| | - Hiroyuki Arai
- Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo 113-0033, Japan
| | - Jun Ogawa
- Division of Applied Life Sciences, Kyoto University Graduate School of Agriculture, Kyoto 606-8502, Japan
| | - Noriyuki Kioka
- Division of Applied Life Sciences, Kyoto University Graduate School of Agriculture, Kyoto 606-8502, Japan
| | - Kazumitsu Ueda
- Division of Applied Life Sciences, Kyoto University Graduate School of Agriculture, Kyoto 606-8502, Japan Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto 606-8502, Japan
| |
Collapse
|
97
|
Abstract
Cholesterol is an essential component for neuronal physiology not only during development stage but also in the adult life. Cholesterol metabolism in brain is independent from that in peripheral tissues due to blood-brain barrier. The content of cholesterol in brain must be accurately maintained in order to keep brain function well. Defects in brain cholesterol metabolism has been shown to be implicated in neurodegenerative diseases, such as Alzheimer's disease (AD), Huntington's disease (HD), Parkinson's disease (PD), and some cognitive deficits typical of the old age. The brain contains large amount of cholesterol, but the cholesterol metabolism and its complex homeostasis regulation are currently poorly understood. This review will seek to integrate current knowledge about the brain cholesterol metabolism with molecular mechanisms.
Collapse
Affiliation(s)
- Juan Zhang
- Chinese Academy of Sciences Key Laboratory of Brain Function and Disease, and School of Life Sciences, University of Science and Technology of China, Hefei, 230026 China
| | - Qiang Liu
- Chinese Academy of Sciences Key Laboratory of Brain Function and Disease, and School of Life Sciences, University of Science and Technology of China, Hefei, 230026 China
| |
Collapse
|
98
|
Saher G, Stumpf SK. Cholesterol in myelin biogenesis and hypomyelinating disorders. Biochim Biophys Acta Mol Cell Biol Lipids 2015; 1851:1083-94. [PMID: 25724171 DOI: 10.1016/j.bbalip.2015.02.010] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 02/05/2015] [Accepted: 02/12/2015] [Indexed: 02/05/2023]
Abstract
The largest pool of free cholesterol in mammals resides in myelin membranes. Myelin facilitates rapid saltatory impulse propagation by electrical insulation of axons. This function is achieved by ensheathing axons with a tightly compacted stack of membranes. Cholesterol influences myelination at many steps, from the differentiation of myelinating glial cells, over the process of myelin membrane biogenesis, to the functionality of mature myelin. Cholesterol emerged as the only integral myelin component that is essential and rate-limiting for the development of myelin in the central and peripheral nervous system. Moreover, disorders that interfere with sterol synthesis or intracellular trafficking of cholesterol and other lipids cause hypomyelination and neurodegeneration. This review summarizes recent results on the roles of cholesterol in CNS myelin biogenesis in normal development and under different pathological conditions. This article is part of a Special Issue entitled Brain Lipids.
Collapse
Affiliation(s)
- Gesine Saher
- Neurogenetics, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany.
| | - Sina Kristin Stumpf
- Neurogenetics, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany.
| |
Collapse
|
99
|
Abstract
Besides their well-documented function of reverse transport of cholesterol, high-density lipoproteins (HDLs) display pleiotropic effects due to their antioxidant, antithrombotic, anti-inflammatory and antiapoptotic properties that may play a major protective role in acute stroke, in particular by limiting the deleterious effects of ischaemia on the blood-brain barrier (BBB) and on the parenchymal cerebral compartment. HDLs may also modulate leukocyte and platelet activation, which may also represent an important target that would justify the use of HDL-based therapy in acute stroke. In this review, we will present an update of all the recent findings in HDL biology that could support a potential clinical use of HDL therapy in ischaemic stroke.
Collapse
|
100
|
Koldamova R, Fitz NF, Lefterov I. ATP-binding cassette transporter A1: from metabolism to neurodegeneration. Neurobiol Dis 2014; 72 Pt A:13-21. [PMID: 24844148 PMCID: PMC4302328 DOI: 10.1016/j.nbd.2014.05.007] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 05/01/2014] [Accepted: 05/06/2014] [Indexed: 01/04/2023] Open
Abstract
ATP-binding cassette transporter A1 (ABCA1) mediates cholesterol efflux to lipid-free apolipoprotein A-I (apoA-I) and apolipoprotein E (apoE). ABCA1 is an essential regulator of high density lipoproteins (HDL) and reverse cholesterol transport - a role that determines its importance for atherosclerosis. Over the last 10 years studies have provided convincing evidence that ABCA1, via its control of apoE lipidation, also has a role in Alzheimer's disease (AD). A series of reports have revealed a significant impact of ABCA1 on Aβ deposition and clearance in AD model mice, as well as an association of common and rare ABCA1 gene variants with the risk for AD. Since APOE is the major genetic risk factor for late onset AD, the regulation of apoE level or its functionality by ABCA1 may prove significant for AD pathogenesis. ABCA1 is transcriptionally regulated by Liver X Receptors (LXR) and Retinoic X Receptors (RXR) which provides a starting point for drug discovery and development of synthetic LXR and RXR agonists for treatment of metabolic and neurodegenerative disorders. This review summarizes the recent results of research on ABCA1, particularly relevant to atherosclerosis and AD.
Collapse
Affiliation(s)
- Radosveta Koldamova
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA 15219, USA.
| | - Nicholas F Fitz
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Iliya Lefterov
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA 15219, USA.
| |
Collapse
|