1
|
Sirtori CR, Cincotto G, Castiglione S, Pavanello C. HDL-replacement therapy: From traditional to emerging clinical applications. ATHEROSCLEROSIS PLUS 2025; 59:68-79. [PMID: 40103705 PMCID: PMC11914826 DOI: 10.1016/j.athplu.2025.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/27/2025] [Accepted: 02/23/2025] [Indexed: 03/20/2025]
Abstract
The unique and multifaceted properties of high-density lipoproteins (HDL)-ranging from cholesterol efflux to anti-inflammatory, anti-oxidant, and immunomodulatory effects-have prompted their direct use, particularly in cardiovascular ischemic conditions. Recent advances have extended the interest in HDL-based treatments to novel applications, from improving stent biocompatibility, to treatment of heart failure to central nervous system (CNS) disorders. Strategies to harness HDL's therapeutic potential have evolved from the direct use of isolated HDL in animal models to reconstituted HDL (rHDL) in humans. For these latter, the use of isolated apoA-I associated with different phospholipids has been the most frequent approach, also involving apparently beneficial mutants, such as the apo A-I Milano (AIM). From the initial very promising results, particularly with this mutant in coronary patients, later studies have mostly been non-confirmatory, although issues such as possible inadequate dose/response and unexpected immunological properties have come to light. Most recently a study on isolated plasma HDL in coronary patients (AEGIS-II) provided overall negative findings, but a clear fall of major cardiovascular events was recorded when restricting analysis to hypercholesterolemic patients. Emerging approaches, including gene therapy and plant-derived recombinant HDL formulations, hold promise for enhancing the accessibility and efficacy of HDL-based interventions. At this time, an improved approach to heart failure treatment also appears feasible, and a better understanding of the role played by HDL in the CNS may lead to significant improvements in the handling of some dramatic diseases at this level. While challenges persist, the evolving landscape of HDL replacement therapies offers hope for significant progress in addressing both cardiovascular and non-cardiovascular conditions.
Collapse
Affiliation(s)
- Cesare Riccardo Sirtori
- Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università Degli Studi di Milano, Milan, Italy
- Dyslipidemia Center, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Giulia Cincotto
- Centro E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università Degli Studi di Milano, Milan, Italy
| | - Sofia Castiglione
- Dyslipidemia Center, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
- Centro E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università Degli Studi di Milano, Milan, Italy
| | - Chiara Pavanello
- Dyslipidemia Center, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
- Centro E. Grossi Paoletti, Dipartimento di Scienze Farmacologiche e Biomolecolari "Rodolfo Paoletti", Università Degli Studi di Milano, Milan, Italy
| |
Collapse
|
2
|
Malajczuk CJ, Mancera RL. Molecular Simulation of the Binding of Amyloid Beta to Apolipoprotein A-I in High-Density Lipoproteins. Int J Mol Sci 2025; 26:1380. [PMID: 39941148 PMCID: PMC11818119 DOI: 10.3390/ijms26031380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/29/2025] [Accepted: 02/05/2025] [Indexed: 02/16/2025] Open
Abstract
Disrupted clearance of amyloid beta (Aβ) from the brain enhances its aggregation and formation of amyloid plaques in Alzheimer's disease. The most abundant protein constituent of circulating high-density lipoprotein (HDL) particles, apoA-I, readily crosses the blood-brain barrier from periphery circulation, exhibits low-micromolar binding affinity for soluble, neurotoxic forms of Aβ, and modulates Aβ aggregation and toxicity in vitro. Its highly conserved N-terminal sequence, 42LNLKLLD48 ('LN'), has been proposed as a binding region for Aβ. However, high-resolution structural characterisation of the mechanism of HDL-Aβ interaction is very difficult to attain. Molecular dynamics simulations were conducted to investigate for the first time the interaction of Aβ and the 'LN' segment of apoA-I. Favourable binding of Aβ by HDLs was found to be driven by hydrophobic and hydrogen-bonding interactions predominantly between the 'LN' segment of apoA-I and Aβ. Preferential binding of Aβ may proceed in small, protein-rich HDLs whereby solvent-exposed hydrophobic 'LN' segments of apoA-I interact specifically with Aβ, stabilising it on the HDL surface in a possibly non-amyloidogenic conformation, facilitating effective Aβ clearance. These findings rationalise the potentially therapeutic role of HDLs in reducing Aβ aggregation and toxicity, and of peptide mimics of the apoA-I interacting region in blocking Aβ aggregation.
Collapse
Affiliation(s)
| | - Ricardo L. Mancera
- Curtin Medical School and Curtin Medical Research Institute, Curtin University, GPO Box U1987, Perth, WA 6845, Australia;
| |
Collapse
|
3
|
Fukuda M, Okanishi H, Ino D, Ono K, Ota T, Wakai E, Sato T, Ohta Y, Kikkawa Y, Inohara H, Kanai Y, Hibino H. Protein profile of mouse endolymph suggests a role in controlling cochlear homeostasis. iScience 2024; 27:111214. [PMID: 39563888 PMCID: PMC11574807 DOI: 10.1016/j.isci.2024.111214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 09/03/2024] [Accepted: 10/16/2024] [Indexed: 11/21/2024] Open
Abstract
The cochlea contains two extracellular fluids, perilymph and endolymph. Endolymph exhibits high potential of approximately +80 to +110 mV (depending on species), which sensitizes sensory hair cells. Other properties of this unique fluid remain elusive, owing to its minuscule volume in rodent cochlea. We therefore developed a technique to collect high-purity endolymph from mouse cochleae. Comprehensive proteomic analysis of sampled endolymph using liquid chromatography with mass spectrometry identified 301 proteins, dominated by molecules engaged in immunity and proteostasis. Approximately 30% of these proteins were undetectable in our perilymph. A combination of mass spectrometry and different approaches revealed that, compared to perilymph, endolymph was enriched with α2-macroglobulin, osteopontin, apolipoprotein D, apolipoprotein E, and apolipoprotein J/clusterin. In other cells or tissues, α2-macroglobulin, apolipoprotein E, and apolipoprotein J contribute to the clearance of degraded proteins from extracellular fluid. Altogether, with the proteins described here, endolymph may play a protective role in stabilizing cochlear homeostasis.
Collapse
Affiliation(s)
- Masatoshi Fukuda
- Division of Glocal Pharmacology, Department of Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
- Department of Otorhinolaryngology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Hiroki Okanishi
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Daisuke Ino
- Division of Glocal Pharmacology, Department of Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Kazuya Ono
- Division of Glocal Pharmacology, Department of Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Takeru Ota
- Division of Glocal Pharmacology, Department of Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Eri Wakai
- Division of Glocal Pharmacology, Department of Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Takashi Sato
- Department of Otorhinolaryngology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yumi Ohta
- Department of Otorhinolaryngology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yoshiaki Kikkawa
- Deafness Project, Department of Basic Medical Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
- Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Japan
- AMED-CREST, AMED, Osaka 565-0871, Japan
| | - Hidenori Inohara
- Department of Otorhinolaryngology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yoshikatsu Kanai
- Department of Bio-system Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
- Institute for Open and Transdisciplinary Research Initiatives, Osaka 565-0871, Japan
- Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Osaka 565-0871, Japan
| | - Hiroshi Hibino
- Division of Glocal Pharmacology, Department of Pharmacology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
- AMED-CREST, AMED, Osaka 565-0871, Japan
| |
Collapse
|
4
|
Wang Z, Zhong R, Curran GL, Min P, Lowe VJ, Li L, Kandimalla KK. High-Density Lipoprotein Mimetic Peptide 4F Reduces Toxic Amyloid-Beta Exposure to the Blood-Brain Barrier Endothelium in Alzheimer's Disease Transgenic Mice. Mol Pharm 2024; 21:5661-5671. [PMID: 39394037 DOI: 10.1021/acs.molpharmaceut.4c00633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2024]
Abstract
Aβ accumulation in the blood-brain barrier (BBB) endothelium, which lines the cerebrovascular lumen, is a significant contributor to cerebrovascular dysfunction in Alzheimer's disease (AD). Reduced high-density lipoprotein (HDL) levels are associated with increased AD risk, and the HDL mimetic peptide 4F has been developed as a promising therapeutic agent to improve cerebrovascular health in AD. In this study, we evaluated the impact of 4F on 125I-Aβ42 blood-to-brain distribution using dynamic SPECT/CT imaging in both wild-type and APP/PS1 transgenic mice. Graphical analysis of the imaging data demonstrated that 4F significantly reduced the blood-to-brain influx rate in wild-type mice and the distribution of 125I-Aβ42 in the BBB endothelium in APP/PS1 mice. To elucidate the molecular mechanisms underlying the effect of 4F, we evaluated its impact on the p38 pathway and its role in mediating Aβ42 trafficking in human BBB endothelial cell monolayers. Treatment with 4F significantly decreased Aβ42 induced p38 activation in BBB endothelial cells. Furthermore, inhibition of p38 kinase significantly reduced endothelial accumulation of fluorescence-labeled Aβ42 and luminal-to-abluminal permeability across the cell monolayer. While our previous publication has hinted at the potential of 4F to reduce Aβ accumulation in the brain parenchyma, the current findings demonstrated the protective effect of 4F in reducing Aβ42 accumulation in the BBB endothelium of AD transgenic mice. These findings revealed the impact of a clinically tested agent, the HDL mimetic peptide 4F, on Aβ exposure to the BBB endothelium and offer novel mechanistic insights into potential therapeutic strategies to treat cerebrovascular dysfunction in AD.
Collapse
Affiliation(s)
- Zengtao Wang
- Department of Pharmaceutics and Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Rui Zhong
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Geoffry L Curran
- Departments of Radiology, Neurology, and Health Sciences, College of Medicine, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Paul Min
- Departments of Radiology, Neurology, and Health Sciences, College of Medicine, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Val J Lowe
- Departments of Radiology, Neurology, and Health Sciences, College of Medicine, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Ling Li
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Karunya K Kandimalla
- Department of Pharmaceutics and Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
5
|
Górska AM, Santos-García I, Eiriz I, Brüning T, Nyman T, Pahnke J. Evaluation of cerebrospinal fluid (CSF) and interstitial fluid (ISF) mouse proteomes for the validation and description of Alzheimer's disease biomarkers. J Neurosci Methods 2024; 411:110239. [PMID: 39102902 DOI: 10.1016/j.jneumeth.2024.110239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 07/22/2024] [Accepted: 07/26/2024] [Indexed: 08/07/2024]
Abstract
BACKGROUND Mass spectrometry (MS)-based cerebrospinal fluid (CSF) proteomics is an important method for discovering biomarkers of neurodegenerative diseases. CSF serves as a reservoir for interstitial fluid (ISF), and extensive communication between the two fluid compartments helps to remove waste products from the brain. NEW METHOD We performed proteomic analyses of both CSF and ISF fluid compartments using intracerebral microdialysis to validate and detect novel biomarkers of Alzheimer's disease (AD) in APPtg and C57Bl/6J control mice. RESULTS We identified up to 625 proteins in ISF and 4483 proteins in CSF samples. By comparing the biofluid profiles of APPtg and C57Bl/6J mice, we detected 37 and 108 significantly up- and downregulated candidates, respectively. In ISF, 7 highly regulated proteins, such as Gfap, Aldh1l1, Gstm1, and Txn, have already been implicated in AD progression, whereas in CSF, 9 out of 14 highly regulated proteins, such as Apba2, Syt12, Pgs1 and Vsnl1, have also been validated to be involved in AD pathogenesis. In addition, we also detected new interesting regulated proteins related to the control of synapses and neurotransmission (Kcna2, Cacng3, and Clcn6) whose roles as AD biomarkers should be further investigated. COMPARISON WITH EXISTING METHODS This newly established combined protocol provides better insight into the mutual communication between ISF and CSF as an analysis of tissue or CSF compartments alone. CONCLUSIONS The use of multiple fluid compartments, ISF and CSF, for the detection of their biological communication enables better detection of new promising AD biomarkers.
Collapse
Affiliation(s)
- Anna Maria Górska
- Translational Neurodegeneration Research and Neuropathology Lab, Department of Clinical Medicine (KlinMed), Medical Faculty, University of Oslo (UiO) and Section of Neuropathology Research, Department of Pathology, Clinics for Laboratory Medicine (KLM), Oslo University Hospital (OUS), Sognsvannsveien 20, Oslo NO-0372, Norway.
| | - Irene Santos-García
- Translational Neurodegeneration Research and Neuropathology Lab, Department of Clinical Medicine (KlinMed), Medical Faculty, University of Oslo (UiO) and Section of Neuropathology Research, Department of Pathology, Clinics for Laboratory Medicine (KLM), Oslo University Hospital (OUS), Sognsvannsveien 20, Oslo NO-0372, Norway.
| | - Ivan Eiriz
- Translational Neurodegeneration Research and Neuropathology Lab, Department of Clinical Medicine (KlinMed), Medical Faculty, University of Oslo (UiO) and Section of Neuropathology Research, Department of Pathology, Clinics for Laboratory Medicine (KLM), Oslo University Hospital (OUS), Sognsvannsveien 20, Oslo NO-0372, Norway.
| | - Thomas Brüning
- Translational Neurodegeneration Research and Neuropathology Lab, Department of Clinical Medicine (KlinMed), Medical Faculty, University of Oslo (UiO) and Section of Neuropathology Research, Department of Pathology, Clinics for Laboratory Medicine (KLM), Oslo University Hospital (OUS), Sognsvannsveien 20, Oslo NO-0372, Norway.
| | - Tuula Nyman
- Proteomics Core Facility, Department of Immunology, Oslo University Hospital (OUS) and University of Oslo (UiO), Faculty of Medicine, Sognsvannsveien 20, Oslo NO-0372, Norway.
| | - Jens Pahnke
- Translational Neurodegeneration Research and Neuropathology Lab, Department of Clinical Medicine (KlinMed), Medical Faculty, University of Oslo (UiO) and Section of Neuropathology Research, Department of Pathology, Clinics for Laboratory Medicine (KLM), Oslo University Hospital (OUS), Sognsvannsveien 20, Oslo NO-0372, Norway; Institute of Nutritional Medicine (INUM) and Lübeck Institute of Dermatology (LIED), University of Lübeck (UzL) and University Medical Center Schleswig-Holstein (UKSH), Ratzeburger Allee 160, Lübeck D-23538, Germany; Department of Pharmacology, Faculty of Medicine and Life Sciences, University of Latvia, Jelgavas iela 3, Rīga LV-1004, Latvia; School of Neurobiology, Biochemistry and Biophysics, The Georg S. Wise Faculty of Life Sciences, Tel Aviv University, Ramat Aviv IL-6997801, Israel.
| |
Collapse
|
6
|
Bonaterra-Pastra A, Solé M, Lope-Piedrafita S, Lucas-Parra M, Castellote L, Marazuela P, Pancorbo O, Rodríguez-Luna D, Hernández-Guillamon M. The presence of circulating human apolipoprotein J reduces the occurrence of cerebral microbleeds in a transgenic mouse model with cerebral amyloid angiopathy. Alzheimers Res Ther 2024; 16:169. [PMID: 39069622 DOI: 10.1186/s13195-024-01541-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND Cerebral amyloid angiopathy (CAA) is characterized by amyloid-β (Aβ) deposition in cerebral vessels, leading to lobar cerebral microbleeds (CMB) and intracerebral hemorrhages (ICH). Apolipoprotein J (ApoJ) is a multifunctional chaperone related to Aβ aggregation and clearance. Our study investigated the vascular impact of chronic recombinant human Apolipoprotein J (rhApoJ) treatment in a transgenic mouse model of β-amyloidosis with prominent CAA. METHODS Twenty-month-old APP23 C57BL/6 mice received 25 doses of rhApoJ (1 mg/kg) (n = 9) or saline (n = 8) intraperitoneally for 13 weeks, while Wild-type (WT) mice received saline (n = 13). Postmortem brains underwent T2*-weighted magnetic resonance imaging (MRI) to detect hemorrhagic lesions. Aβ levels and distribution, cerebral fibrinogen leakage, brain smooth muscle actin (sma), and plasma matrix metalloproteinases and inflammatory markers were analyzed after treatments. Additionally, plasma samples from 22 patients with lobar ICH were examined to determine the clinical relevance of the preclinical findings. RESULTS rhApoJ-treated APP23 presented fewer cortical CMBs (50-300 μm diameter) (p = 0.012) and cortical larger hemorrhages (> 300 μm) (p = 0.002) than saline-treated mice, independently of Aβ brain levels. MRI-detected hemorrhagic lesions correlated with fibrinogen cerebral extravasation (p = 0.011). Additionally, rhApoJ-treated mice presented higher number of sma-positive vessels than saline-treated mice (p = 0.038). In rhApoJ-treated mice, human ApoJ was detected in plasma and in occasional leptomeningeal vessels, but not in the parenchyma, suggesting that its mechanism of action operates through the periphery. The administration of rhApoJ induced an increase in plasma Groα (p = 0.035) and MIP-1α (p = 0.035) levels, while lower MMP-12 (p = 0.046) levels, compared to the saline-treated group. In acute lobar ICH patients, MMP-12 plasma levels correlated with larger hemorrhage volume (p = 0.040) and irregular ICH shape (p = 0.036). CONCLUSIONS Chronic rhApoJ treatment in aged APP23 mice ameliorated CAA-related neurovascular damage by reducing the occurrence of CMB. We propose that rhApoJ may prevent blood-brain barrier (BBB) leakage and CMB appearance partly through circulating MMP-12 modulation.
Collapse
Affiliation(s)
- Anna Bonaterra-Pastra
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Passeig Vall d'Hebron, 119-129, Mediterranean Building, 1st floor, lab 106, Barcelona, 08035, Spain
| | - Montse Solé
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Passeig Vall d'Hebron, 119-129, Mediterranean Building, 1st floor, lab 106, Barcelona, 08035, Spain
- Department of Bioquímica i Biologia Molecular i Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra (Barcelona), Spain
| | - Silvia Lope-Piedrafita
- Nuclear Magnetic Resonance Service, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Maria Lucas-Parra
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Passeig Vall d'Hebron, 119-129, Mediterranean Building, 1st floor, lab 106, Barcelona, 08035, Spain
| | - Laura Castellote
- Department of Clinical Biochemistry, Clinical Laboratories, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Paula Marazuela
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Passeig Vall d'Hebron, 119-129, Mediterranean Building, 1st floor, lab 106, Barcelona, 08035, Spain
| | - Olalla Pancorbo
- Stroke Research Group, Vall d'Hebron Research Institute, Barcelona, Spain
| | | | - Mar Hernández-Guillamon
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Passeig Vall d'Hebron, 119-129, Mediterranean Building, 1st floor, lab 106, Barcelona, 08035, Spain.
| |
Collapse
|
7
|
Smith AS, Subramanian J, Doderer J, Moskovitz J. Methionine oxidation of clusterin in Alzheimer's disease and its effect on clusterin's binding to beta-amyloid. Neurosci Lett 2024; 836:137874. [PMID: 38857696 DOI: 10.1016/j.neulet.2024.137874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 03/20/2024] [Accepted: 06/07/2024] [Indexed: 06/12/2024]
Abstract
Clusterin is a secreted glycoprotein that participates in multiple physiological processes through its chaperon function. In Alzheimer's disease, the brain functions under an increased oxidative stress condition that causes an elevation of protein oxidation, resulting in enhanced pathology. Accordingly, it is important to determine the type of human brain cells that are mostly prone to methionine oxidation in Alzheimer's disease and specifically monitoring the methionine-oxidation levels of clusterin in human and mice brains and its effect on clusterin's function. We analyzed the level of methionine sulfoxide (MetO)-clusterin in these brains, using a combination of immunoprecipitation and Western-blott analyses. Also, we determine the effect of methionine oxidation on clusterin ability to bind beta-amyloid, in vitro, using calorimetric assay. Our results show that human neurons and astrocytes of Alzheimer's disease brains are mostly affected by methionine oxidation. Moreover, MetO-clusterin levels are elevated in postmortem Alzheimer's disease human and mouse brains in comparison to controls. Finally, oxidation of methionine residues of purified clusterin reduced its binding efficiency to beta-amyloid. In conclusion, we suggest that methionine oxidation of brain-clusterin is enhanced in Alzheimer's disease and that this oxidation compromises its chaperon function, leading to exacerbation of beta-amyloid's toxicity in Alzheimer's disease.
Collapse
Affiliation(s)
- Adam S Smith
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
| | - Jaichandar Subramanian
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
| | - Julia Doderer
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
| | - Jackob Moskovitz
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA.
| |
Collapse
|
8
|
Lan X, Qin S, Liu H, Guo M, Zhang Y, Jin X, Duan X, Sun M, Liu Z, Wang W, Zheng Q, Liao X, Chen J, Kang Y, Xie Y, Song X. Dual-targeting tigecycline nanoparticles for treating intracranial infections caused by multidrug-resistant Acinetobacter baumannii. J Nanobiotechnology 2024; 22:138. [PMID: 38555444 PMCID: PMC10981309 DOI: 10.1186/s12951-024-02373-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 02/26/2024] [Indexed: 04/02/2024] Open
Abstract
Multidrug-resistant (MDR) Acinetobacter baumannii (A. baumannii) is a formidable pathogen responsible for severe intracranial infections post-craniotomy, exhibiting a mortality rate as high as 71%. Tigecycline (TGC), a broad-spectrum antibiotic, emerged as a potential therapeutic agent for MDR A. baumannii infections. Nonetheless, its clinical application was hindered by a short in vivo half-life and limited permeability through the blood-brain barrier (BBB). In this study, we prepared a novel core-shell nanoparticle encapsulating water-soluble tigecycline using a blend of mPEG-PLGA and PLGA materials. This nanoparticle, modified with a dual-targeting peptide Aβ11 and Tween 80 (Aβ11/T80@CSs), was specifically designed to enhance the delivery of tigecycline to the brain for treating A. baumannii-induced intracranial infections. Our findings demonstrated that Aβ11/T80@CSs nanocarriers successfully traversed the BBB and effectively delivered TGC into the cerebrospinal fluid (CSF), leading to a significant therapeutic response in a model of MDR A. baumannii intracranial infection. This study offers initial evidence and a platform for the application of brain-targeted nanocarrier delivery systems, showcasing their potential in administering water-soluble anti-infection drugs for intracranial infection treatments, and suggesting promising avenues for clinical translation.
Collapse
Affiliation(s)
- Xing Lan
- Department of Critical Care Medicine, Department of Clinical Pharmacy, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China School of Nursing, West China Hospital, Sichuan University, Chengdu, China
- State Key Laboratory of Drug Delivery and Pharmacokinetics, Tianjin Institute of Pharmaceutical Research, Tianjin, 300301, People's Republic of China
| | - Shugang Qin
- Department of Critical Care Medicine, Department of Clinical Pharmacy, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China School of Nursing, West China Hospital, Sichuan University, Chengdu, China
| | - Huan Liu
- Department of Critical Care Medicine, Department of Clinical Pharmacy, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China School of Nursing, West China Hospital, Sichuan University, Chengdu, China
| | - Mengran Guo
- Department of Critical Care Medicine, Department of Clinical Pharmacy, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China School of Nursing, West China Hospital, Sichuan University, Chengdu, China
| | - Yupei Zhang
- Department of Critical Care Medicine, Department of Clinical Pharmacy, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China School of Nursing, West China Hospital, Sichuan University, Chengdu, China
| | - Xinyang Jin
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau, China
- Shihezi University, Xinjiang, China
| | - Xing Duan
- Department of Critical Care Medicine, Department of Clinical Pharmacy, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China School of Nursing, West China Hospital, Sichuan University, Chengdu, China
| | - Min Sun
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau, China
- Shihezi University, Xinjiang, China
| | - Zhenjun Liu
- Department of Critical Care Medicine, Department of Clinical Pharmacy, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China School of Nursing, West China Hospital, Sichuan University, Chengdu, China
| | - Wenyan Wang
- Department of Critical Care Medicine, Department of Clinical Pharmacy, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China School of Nursing, West China Hospital, Sichuan University, Chengdu, China
| | - Qian Zheng
- Department of Critical Care Medicine, Department of Clinical Pharmacy, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China School of Nursing, West China Hospital, Sichuan University, Chengdu, China
| | - Xuelian Liao
- Department of Critical Care Medicine, Department of Clinical Pharmacy, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China School of Nursing, West China Hospital, Sichuan University, Chengdu, China
| | - Jinpeng Chen
- State Key Laboratory of Drug Delivery and Pharmacokinetics, Tianjin Institute of Pharmaceutical Research, Tianjin, 300301, People's Republic of China
| | - Yan Kang
- Department of Critical Care Medicine, Department of Clinical Pharmacy, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China School of Nursing, West China Hospital, Sichuan University, Chengdu, China.
| | - Yongmei Xie
- Department of Critical Care Medicine, Department of Clinical Pharmacy, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China School of Nursing, West China Hospital, Sichuan University, Chengdu, China.
| | - Xiangrong Song
- Department of Critical Care Medicine, Department of Clinical Pharmacy, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China School of Nursing, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
9
|
Saheli M, Moshrefi M, Baghalishahi M, Mohkami A, Firouzi Y, Suzuki K, Khoramipour K. Cognitive Fitness: Harnessing the Strength of Exerkines for Aging and Metabolic Challenges. Sports (Basel) 2024; 12:57. [PMID: 38393277 PMCID: PMC10891799 DOI: 10.3390/sports12020057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/31/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Addressing cognitive impairment (CI) represents a significant global challenge in health and social care. Evidence suggests that aging and metabolic disorders increase the risk of CI, yet promisingly, physical exercise has been identified as a potential ameliorative factor. Specifically, there is a growing understanding that exercise-induced cognitive improvement may be mediated by molecules known as exerkines. This review delves into the potential impact of aging and metabolic disorders on CI, elucidating the mechanisms through which various exerkines may bolster cognitive function in this context. Additionally, the discussion extends to the role of exerkines in facilitating stem cell mobilization, offering a potential avenue for improving cognitive impairment.
Collapse
Affiliation(s)
- Mona Saheli
- Department of Anatomical Sciences, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman 7616913555, Iran; (M.S.); (M.B.)
| | - Mandana Moshrefi
- Department of Physiology and Pharmacology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman 7616913555, Iran;
| | - Masoumeh Baghalishahi
- Department of Anatomical Sciences, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman 7616913555, Iran; (M.S.); (M.B.)
| | - Amirhossein Mohkami
- Department of Exercise Physiology, Faculty of Sport Sciences, Hakim Sabzevari University, Sabzevar 9617976487, Iran;
| | - Yaser Firouzi
- Department of Exercise Physiology, Faculty of Sport Sciences, Shahid Bahonar University, Kerman 7616913439, Iran;
| | - Katsuhiko Suzuki
- Faculty of Sport Sciences, Waseda University, Tokorozawa 359-1192, Japan
| | - Kayvan Khoramipour
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman 7619813159, Iran
| |
Collapse
|
10
|
Liang J, LaFleur B, Hussainy S, Perry G. Gene Co-Expression Analysis of Multiple Brain Tissues Reveals Correlation of FAM222A Expression with Multiple Alzheimer's Disease-Related Genes. J Alzheimers Dis 2024; 99:S249-S263. [PMID: 37092222 PMCID: PMC11091573 DOI: 10.3233/jad-221241] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2023] [Indexed: 04/25/2023]
Abstract
Background Alzheimer's disease (AD) is the most common form of dementia in the elderly marked by central nervous system (CNS) neuronal loss and amyloid plaques. FAM222A, encoding an amyloid plaque core protein, is an AD brain atrophy susceptibility gene that mediates amyloid-β aggregation. However, the expression interplay between FAM222A and other AD-related pathway genes is unclear. Objective Our goal was to study FAM222A's whole-genome co-expression profile in multiple tissues and investigate its interplay with other AD-related genes. Methods We analyzed gene expression correlations in Genotype-Tissue Expression (GTEx) tissues to identify FAM222A co-expressed genes and performed functional enrichment analysis on identified genes in CNS system. Results Genome-wide gene expression profiling identified 673 genes significantly correlated with FAM222A (p < 2.5×10-6) in 48 human tissues, including 298 from 13 CNS tissues. Functional enrichment analysis revealed that FAM222A co-expressed CNS genes were enriched in multiple AD-related pathways. Gene co-expression network analysis for identified genes in each brain region predicted other disease associated genes with similar biological function. Furthermore, co-expression of 25 out of 31 AD-related pathways genes with FAM222A was replicated in brain samples from 107 aged subjects from the Aging, Dementia and TBI Study. Conclusion This gene co-expression study identified multiple AD-related genes that are associated with FAM222A, indicating that FAM222A and AD-associated genes can be active simultaneously in similar biological processes, providing evidence that supports the association of FAM222A with AD.
Collapse
Affiliation(s)
- Jingjing Liang
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Arizona, Tucson, AZ, USA
| | - Bonnie LaFleur
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Arizona, Tucson, AZ, USA
| | - Sadiya Hussainy
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Arizona, Tucson, AZ, USA
| | - George Perry
- College of Sciences, University of Texas at San Antonio, San Antonio, TX, USA
| |
Collapse
|
11
|
Zhou M, Fu X, Ma B, Chen Z, Cheng Y, Liu L, Kan S, Zhao X, Feng S, Jiang Z, Zhu R. Effects of low-intensity ultrasound opening the blood-brain barrier on Alzheimer's disease-a mini review. Front Neurol 2023; 14:1274642. [PMID: 38020620 PMCID: PMC10646525 DOI: 10.3389/fneur.2023.1274642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 09/29/2023] [Indexed: 12/01/2023] Open
Abstract
Due to the complex pathological mechanisms of Alzheimer's disease (AD), its treatment remains a challenge. One of the major difficulties in treating AD is the difficulty for drugs to cross the blood-brain barrier (BBB). Low-intensity ultrasound (LIUS) is a novel type of ultrasound with neuromodulation function. It has been widely reported that LIUS combined with intravenous injection of microbubbles (MB) can effectively, safely, and reversibly open the BBB to achieve non-invasive targeted drug delivery. However, many studies have reported that LIUS combined with MB-mediated BBB opening (LIUS + MB-BBBO) can improve pathological deposition and cognitive impairment in AD patients and mice without delivering additional drugs. This article reviews the relevant research studies on LIUS + MB-BBBO in the treatment of AD, analyzes its potential mechanisms, and summarizes relevant ultrasound parameters.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Rusen Zhu
- Tianjin Union Medical Center, Tianjin, China
| |
Collapse
|
12
|
Wang Q, Huang X, Su Y, Yin G, Wang S, Yu B, Li H, Qi J, Chen H, Zeng W, Zhang K, Verkhratsky A, Niu J, Yi C. Activation of Wnt/β-catenin pathway mitigates blood-brain barrier dysfunction in Alzheimer's disease. Brain 2022; 145:4474-4488. [PMID: 35788280 DOI: 10.1093/brain/awac236] [Citation(s) in RCA: 98] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 05/29/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that causes age-dependent neurological and cognitive declines. The treatments for AD pose a significant challenge, because the mechanisms of disease are not being fully understood. Malfunction of the blood-brain barrier (BBB) is increasingly recognized as a major contributor to the pathophysiology of AD, especially at the early stages of the disease. However, the underlying mechanisms remain poorly characterized, while few molecules can directly target and improve BBB function in the context of AD. Here, we showed dysfunctional BBB in AD patients reflected by perivascular accumulation of blood-derived fibrinogen in the hippocampus and cortex, accompanied by decreased tight junction proteins Claudin-5 and glucose transporter Glut-1 in the brain endothelial cells (BECs). In the APPswe/PS1dE9 (APP/PS1) mouse model of AD, BBB dysfunction started at 4 months of age and became severe at 9 months of age. In the cerebral microvessels of APP/PS1 mice and Aβ-treated BECs, we found suppressed Wnt/β-catenin signaling triggered by an increase of GSK3β activation, but not an inhibition of the AKT pathway or switching to the Wnt/planar cell polarity pathway. Furthermore, using our newly developed optogenetic tool for controlled regulation of LRP6 (upstream regulator of the Wnt signaling) to activate Wnt/β-catenin pathway, BBB malfunction was restored by preventing Aβ-induced BEC impairments and promoting the barrier repair. In conclusion, targeting LRP6 in the Wnt/β-catenin pathway in the brain endothelium can alleviate BBB malfunction induced by Aβ, which may be a potential treatment strategy for AD.
Collapse
Affiliation(s)
- Qi Wang
- Research Centre, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China.,Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing, China
| | - Xiaomin Huang
- Research Centre, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Yixun Su
- Research Centre, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China.,Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing, China
| | - Guowei Yin
- Research Centre, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Shouyu Wang
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing, China
| | - Bin Yu
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing, China
| | - Hui Li
- Research Centre, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China.,Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing, China
| | - Junhua Qi
- Research Centre, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Hui Chen
- School of Life Sciences, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Wen Zeng
- Department of Cell Biology, State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University, Chongqing, China
| | - Kai Zhang
- Department of Biochemistry, School of Molecular and Cellular Biology, the University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.,Achucarro Center for Neuroscience, IKERBASQUE, Basque Foundation for Science, 48011 Bilbao, Spain
| | - Jianqin Niu
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing, China
| | - Chenju Yi
- Research Centre, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
13
|
Tong JH, Gong SQ, Zhang YS, Dong JR, Zhong X, Wei MJ, Liu MY. Association of Circulating Apolipoprotein AI Levels in Patients With Alzheimer's Disease: A Systematic Review and Meta-Analysis. Front Aging Neurosci 2022; 14:899175. [PMID: 35663584 PMCID: PMC9157647 DOI: 10.3389/fnagi.2022.899175] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 04/19/2022] [Indexed: 11/26/2022] Open
Abstract
With the development of medicine, our research on Alzheimer's disease (AD) has been further deepened, but the mechanism of its occurrence and development has not been fully revealed, and there is currently no effective treatment method. Several studies have shown that apolipoprotein AI (ApoA-I) can affect the occurrence and development of Alzheimer's disease by binding to amyloid β (Aβ). However, the association between circulating levels of ApoA-I and AD remains controversial. We conducted a meta-analysis of 18 studies published between 1992 and 2017 to determine whether the ApoA-I levels in the blood and cerebrospinal fluid (CSF) are abnormal in AD. Literatures were searched in PubMed, EMBASE and Web of Science databases without language limitations. A pooled subject sample including 1,077 AD patients and 1,271 healthy controls (HCs) was available to assess circulating ApoA-I levels; 747 AD patients and 680 HCs were included for ApoA-I levels in serum; 246 AD patients and 456 HCs were included for ApoA-I levels in plasma; 201 AD patients and 447 HCs were included for ApoA-I levels in CSF. It was found that serum and plasma levels of ApoA-I were significantly reduced in AD patients compared with HCs {[standardized mean difference (SMD) = −1.16; 95% confidence interval (CI) (−1.72, −0.59); P = 0.000] and [SMD = −1.13; 95% CI (−2.05, −0.21); P = 0.016]}. Patients with AD showed a tendency toward higher CSF ApoA-I levels compared with HCs, although this difference was non-significant [SMD = 0.20; 95% CI (−0.16, 0.56); P = 0.273]. In addition, when we analyzed the ApoA-I levels of serum and plasma together, the circulating ApoA-I levels in AD patients was significantly lower [SMD = −1.15; 95% CI (−1.63, −0.66); P = 0.000]. These results indicate that ApoA-I deficiency may be a risk factor of AD, and ApoA-I has the potential to serve as a biomarker for AD and provide experimental evidence for diagnosis of AD. Systematic Review Registration: PROSPERO, identifier: 325961.
Collapse
|
14
|
Vargas-George S, Dave KR. Models of cerebral amyloid angiopathy-related intracerebral hemorrhage. BRAIN HEMORRHAGES 2022. [DOI: 10.1016/j.hest.2022.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
15
|
Berdowska I, Matusiewicz M, Krzystek-Korpacka M. HDL Accessory Proteins in Parkinson’s Disease—Focusing on Clusterin (Apolipoprotein J) in Regard to Its Involvement in Pathology and Diagnostics—A Review. Antioxidants (Basel) 2022; 11:antiox11030524. [PMID: 35326174 PMCID: PMC8944556 DOI: 10.3390/antiox11030524] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/01/2022] [Accepted: 03/03/2022] [Indexed: 02/04/2023] Open
Abstract
Parkinson’s disease (PD)—a neurodegenerative disorder (NDD) characterized by progressive destruction of dopaminergic neurons within the substantia nigra of the brain—is associated with the formation of Lewy bodies containing mainly α-synuclein. HDL-related proteins such as paraoxonase 1 and apolipoproteins A1, E, D, and J are implicated in NDDs, including PD. Apolipoprotein J (ApoJ, clusterin) is a ubiquitous, multifunctional protein; besides its engagement in lipid transport, it modulates a variety of other processes such as immune system functionality and cellular death signaling. Furthermore, being an extracellular chaperone, ApoJ interacts with proteins associated with NDD pathogenesis (amyloid β, tau, and α-synuclein), thus modulating their properties. In this review, the association of clusterin with PD is delineated, with respect to its putative involvement in the pathological mechanism and its application in PD prognosis/diagnosis.
Collapse
Affiliation(s)
- Izabela Berdowska
- Correspondence: (I.B.); (M.M.); Tel.: +48-71-784-13-92 (I.B.); +48-71-784-13-70 (M.M.)
| | | | | |
Collapse
|
16
|
Ding Y, Shusta EV, Palecek SP. Integrating in vitro disease models of the neurovascular unit into discovery and development of neurotherapeutics. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2021; 20:100341. [PMID: 34693102 PMCID: PMC8530278 DOI: 10.1016/j.cobme.2021.100341] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The blood-brain barrier (BBB) regulates the transport of small molecules, proteins, and cells between the bloodstream and the central nervous system (CNS). Brain microvascular endothelial cells work with other resident brain cell types, including pericytes, astrocytes, neurons, and microglia, to form the neurovascular unit (NVU) and maintain BBB integrity. The restrictive barrier influences the pathogenesis of many CNS diseases, and impedes the delivery of neurotherapeutics into the CNS. In vitro NVU models enable the discovery of complex cell-cell interactions involved in human BBB pathophysiology in diseases including Alzheimer's Disease (AD), Parkinson's Disease (PD) and viral infections of the brain. In vitro NVU models have also been deployed to study the delivery of neurotherapeutics across the BBB, including small molecule drugs, monoclonal antibodies, gene therapy vectors and immune cells. The high scalability, accessibility, and phenotype fidelity of in vitro NVU models can facilitate the discovery and development of effective neurotherapeutics.
Collapse
Affiliation(s)
- Yunfeng Ding
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Eric V Shusta
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, USA
- Department of Neurological Surgery, University of Wisconsin-Madison, Madison, WI, USA
| | - Sean P Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
17
|
Uddin MS, Kabir MT, Begum MM, Islam MS, Behl T, Ashraf GM. Exploring the Role of CLU in the Pathogenesis of Alzheimer's Disease. Neurotox Res 2021; 39:2108-2119. [PMID: 32820456 DOI: 10.1007/s12640-020-00271-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 08/05/2020] [Accepted: 08/10/2020] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) is a chronic and devastating neurodegenerative disorder that is affecting elderly people at an increasing rate. Clusterin (CLU), an extracellular chaperone, is an ubiquitously expressed protein that can be identified in various body fluids and tissues. Expression of CLU can lead to various processes including suppression of complement system, lipid transport, chaperone function, and also controlling neuronal cell death and cell survival mechanisms. Studies have confirmed that the level of CLU expression is increased in AD. Furthermore, CLU also decreased the toxicity and aggregation of amyloid beta (Aβ). However when the Aβ level was far greater than CLU, then the amyloid generation was increased. CLU was also found to incorporate in the amyloid aggregates, which were more harmful as compared with the Aβ42 aggregates alone. Growing evidence indicates that CLU plays roles in AD pathogenesis via various processes, including aggregation and clearance of Aβ, neuroinflammation, lipid metabolism, Wnt signaling, copper homeostasis, and regulation of neuronal cell cycle and apoptosis. In this article, we represent the critical interaction of CLU and AD based on recent advances. Furthermore, we have also focused on the Aβ-dependent and Aβ-independent mechanisms by which CLU plays a role in AD pathogenesis.
Collapse
Affiliation(s)
- Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh.
- Pharmakon Neuroscience Research Network, Dhaka, Bangladesh.
| | | | | | | | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
18
|
Kurz C, Walker L, Rauchmann BS, Perneczky R. Dysfunction of the blood-brain barrier in Alzheimer's disease: evidence from human studies. Neuropathol Appl Neurobiol 2021; 48:e12782. [PMID: 34823269 DOI: 10.1111/nan.12782] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 11/04/2021] [Accepted: 11/16/2021] [Indexed: 11/28/2022]
Abstract
The pathological processes leading to synapse loss, neuronal loss, brain atrophy and gliosis in Alzheimer´s disease (AD) and their relation to vascular disease and immunological changes are yet to be fully explored. Amyloid-β (Aβ) aggregation, vascular damage and altered immune response interact at the blood-brain-barrier (BBB), affecting the brain endothelium and fuelling neurodegeneration. The aim of the present systematic literature review was to critically appraise and to summarise the published evidence on the clinical correlations and pathophysiological concepts of BBB damage in AD, focusing on human data. The PubMed, Cochrane, Medline and Embase databases were searched for original research articles, systematic reviews and meta-analyses, published in English language from 01/2000 to 07/2021, using the keywords Alzheimer*, amyloid-β or β-amyloid or abeta and brain-blood barrier or BBB. This review shows that specific changes of intercellular structures, reduced expression of transendothelial carriers, induction of vasoactive mediators and activation of both astroglia and monocytes/macrophages characterise blood-brain barrier damage in human AD and AD models. BBB dysfunction on magnetic resonance imaging takes place early in the disease course in AD-specific brain regions. The toxic effects of Aβ and apolipoprotein E (ApoE) are likely to induce a non-cerebral-amyloid-angiopathy-related degeneration of endothelial cells, independently of cerebrovascular disease; however, some of the observed structural changes may just arise with age. Small vessel disease, ApoE, loss of pericytes, pro-inflammatory signalling and cerebral amyloid angiopathy enhance blood-brain-barrier damage. Novel therapeutic approaches for AD, including magnetic resonance-guided focused ultrasound, aim to open the BBB, potentially leading to an improved drainage of Aβ along perivascular channels and increased elimination from the brain. In vitro treatments with ApoE-modifying agents yielded promising effects on modulating BBB function. Reducing cardiovascular risk factors represents one of the most promising interventions for dementia prevention at present. However, further research is needed to elucidate the connection of BBB damage and tau pathology, the role of pro-inflammatory mediators in draining macromolecules and cells from the cerebral parenchyma, including their contribution to cerebral amyloid angiopathy. Improved insight into these pathomechanisms may allow to shed light on the role of Aβ deposition as a primary vs. a secondary event in the complex pathogenesis of AD.
Collapse
Affiliation(s)
- Carolin Kurz
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
| | - Lauren Walker
- Translational and Clinical Research Institute, Campus for Ageing and Vitality, Newcastle University
| | - Boris-Stephan Rauchmann
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany.,Department of Radiology, Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Robert Perneczky
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany.,German Center for Neurodegenerative Disorders (DZNE) Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College London, London, UK
| |
Collapse
|
19
|
Rostagno A, Calero M, Holton JL, Revesz T, Lashley T, Ghiso J. Association of clusterin with the BRI2-derived amyloid molecules ABri and ADan. Neurobiol Dis 2021; 158:105452. [PMID: 34298087 PMCID: PMC8440498 DOI: 10.1016/j.nbd.2021.105452] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 06/30/2021] [Accepted: 07/18/2021] [Indexed: 10/20/2022] Open
Abstract
Familial British and Danish dementias (FBD and FDD) share striking neuropathological similarities with Alzheimer's disease (AD), including intraneuronal neurofibrillary tangles as well as parenchymal and vascular amyloid deposits. Multiple amyloid associated proteins with still controversial role in amyloidogenesis colocalize with the structurally different amyloid peptides ABri in FBD, ADan in FDD, and Aβ in AD. Genetic variants and plasma levels of one of these associated proteins, clusterin, have been identified as risk factors for AD. Clusterin is known to bind soluble Aβ in biological fluids, facilitate its brain clearance, and prevent its aggregation. The current work identifies clusterin as the major ABri- and ADan-binding protein and provides insight into the biochemical mechanisms leading to the association of clusterin with ABri and ADan deposits. Mirroring findings in AD, the studies corroborate clusterin co-localization with cerebral parenchymal and vascular amyloid deposits in both disorders. Ligand affinity chromatography with downstream Western blot and amino acid sequence analyses unequivocally identified clusterin as the major ABri- and ADan-binding plasma protein. ELISA highlighted a specific saturable binding of clusterin to ABri and ADan with low nanomolar Kd values within the same range as those previously demonstrated for the clusterin-Aβ interaction. Consistent with its chaperone activity, thioflavin T binding assays clearly showed a modulatory effect of clusterin on ABri and ADan aggregation/fibrillization properties. Our findings, together with the known multifunctional activity of clusterin and its modulatory activity on the complex cellular pathways leading to oxidative stress, mitochondrial dysfunction, and the induction of cell death mechanisms - all known pathogenic features of these protein folding disorders - suggests the likelihood of a more complex role and a translational potential for the apolipoprotein in the amelioration/prevention of these pathogenic mechanisms.
Collapse
Affiliation(s)
- Agueda Rostagno
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Miguel Calero
- Instituto de Salud Carlos III, 28029 Madrid, Spain; Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), 28031 Madrid, Spain; Alzheimer's Center Reina Sofia Foundation - CIEN Foundation, 28031 Madrid, Spain
| | - Janice L Holton
- The Queen Square Brain Bank for Neurological Disorders, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Tamas Revesz
- The Queen Square Brain Bank for Neurological Disorders, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK; Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Tammaryn Lashley
- The Queen Square Brain Bank for Neurological Disorders, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK; Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Jorge Ghiso
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA; Department of Psychiatry, New York University School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
20
|
Van Valkenburgh J, Meuret C, Martinez AE, Kodancha V, Solomon V, Chen K, Yassine HN. Understanding the Exchange of Systemic HDL Particles Into the Brain and Vascular Cells Has Diagnostic and Therapeutic Implications for Neurodegenerative Diseases. Front Physiol 2021; 12:700847. [PMID: 34552500 PMCID: PMC8450374 DOI: 10.3389/fphys.2021.700847] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 07/29/2021] [Indexed: 12/02/2022] Open
Abstract
High-density lipoproteins (HDLs) are complex, heterogenous lipoprotein particles, consisting of a large family of apolipoproteins, formed in subspecies of distinct shapes, sizes, and functions and are synthesized in both the brain and the periphery. HDL apolipoproteins are important determinants of Alzheimer’s disease (AD) pathology and vascular dementia, having both central and peripheral effects on brain amyloid-beta (Aβ) accumulation and vascular functions, however, the extent to which HDL particles (HLD-P) can exchange their protein and lipid components between the central nervous system (CNS) and the systemic circulation remains unclear. In this review, we delineate how HDL’s structure and composition enable exchange between the brain, cerebrospinal fluid (CSF) compartment, and vascular cells that ultimately affect brain amyloid metabolism and atherosclerosis. Accordingly, we then elucidate how modifications of HDL-P have diagnostic and therapeutic potential for brain vascular and neurodegenerative diseases.
Collapse
Affiliation(s)
- Juno Van Valkenburgh
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Cristiana Meuret
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Ashley E Martinez
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Vibha Kodancha
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Victoria Solomon
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Kai Chen
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Hussein N Yassine
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
21
|
Jin Y, Chifodya K, Han G, Jiang W, Chen Y, Shi Y, Xu Q, Xi Y, Wang J, Zhou J, Zhang H, Ding Y. High-density lipoprotein in Alzheimer's disease: From potential biomarkers to therapeutics. J Control Release 2021; 338:56-70. [PMID: 34391838 DOI: 10.1016/j.jconrel.2021.08.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 08/09/2021] [Accepted: 08/10/2021] [Indexed: 12/17/2022]
Abstract
The inverse correlation between high-density lipoprotein (HDL) levels in vivo and the risk of Alzheimer's disease (AD) has become an inspiration for HDL-inspired AD therapy, including plain HDL and various intelligent HDL-based drug delivery systems. In this review, we will focus on the two endogenous HDL subtypes in the central nervous system (CNS), apolipoprotein E-based HDL (apoE-HDL) and apolipoprotein A-I-based HDL (apoA-I-HDL), especially their influence on AD pathophysiology to reveal HDL's potential as biomarkers for risk prediction, and summarize the relevant therapeutic mechanisms to propose possible treatment strategies. We will emphasize the latest advances of HDL as therapeutics (plain HDL and HDL-based drug delivery systems) to discuss the potential for AD therapy and review innovative techniques in the preparation of HDL-based nanoplatforms to provide a basis for the rational design and future development of anti-AD drugs.
Collapse
Affiliation(s)
- Yi Jin
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Ministry of Education, Nanjing 210009, China; State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China; NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, Nanjing 210009, China
| | - Kudzai Chifodya
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Guochen Han
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Ministry of Education, Nanjing 210009, China; State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China; NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, Nanjing 210009, China
| | - Wenxin Jiang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Yun Chen
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Yang Shi
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Qiao Xu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Yilong Xi
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Jun Wang
- Department of Geriatrics, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Jianping Zhou
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Ministry of Education, Nanjing 210009, China; State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China; NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, Nanjing 210009, China.
| | - Huaqing Zhang
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Ministry of Education, Nanjing 210009, China; State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China; NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, Nanjing 210009, China.
| | - Yang Ding
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Ministry of Education, Nanjing 210009, China; State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China; NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, Nanjing 210009, China.
| |
Collapse
|
22
|
Zuin M, Cervellati C, Trentini A, Passaro A, Rosta V, Zimetti F, Zuliani G. Association between Serum Concentrations of Apolipoprotein A-I (ApoA-I) and Alzheimer's Disease: Systematic Review and Meta-Analysis. Diagnostics (Basel) 2021; 11:984. [PMID: 34071695 PMCID: PMC8229134 DOI: 10.3390/diagnostics11060984] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/26/2021] [Accepted: 05/27/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND A wealth of experimental and epidemiological evidence suggest that Apolipoprotein A-I (ApoA-I), the main protein constituent of high-density lipoprotein (HDL), may protect against Alzheimer disease (AD). To investigate this potential role, we conducted a meta-analysis of the published studies on the relationship between serum ApoA-I and AD occurrence. METHODS We screened MEDLINE, EMBASE, Web of Science, and Scopus, for cross-sectional studies published from inception to 1 March 2021, comparing the ApoA-I serum levels between patients with AD and cognitively normal controls. RESULTS From an initial screening of 245 articles, 5 studies, including 397 AD patients (mean age 75.0 years, 234 females) and 367 controls (mean age 69.2 years, 182 females), met the inclusion criteria. Compared to healthy controls, AD subjects had a lower ApoA-I serum level. The pooled weighted mean difference from a random-effects model was -0.31 g/L (p < 0.0001) (95% Confidence Interval: [-0.62-0.01], with high heterogeneity (I2 = 100%). The Egger's test confirmed an absence of publication bias (t = 0.62, p = 0.576). CONCLUSIONS Our study showed that AD patients present lower serum levels of ApoA-I compared to cognitively normal individuals. Further studies on large population samples are required to support this finding.
Collapse
Affiliation(s)
- Marco Zuin
- Department of Translational Medicine and for Romagna, University of Ferrara, 44121 Ferrara, Italy
| | - Carlo Cervellati
- Department of Translational Medicine and for Romagna, University of Ferrara, 44121 Ferrara, Italy
| | - Alessandro Trentini
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Angelina Passaro
- Department of Translational Medicine and for Romagna, University of Ferrara, 44121 Ferrara, Italy
| | - Valentina Rosta
- Department of Translational Medicine and for Romagna, University of Ferrara, 44121 Ferrara, Italy
| | - Francesca Zimetti
- Department of Food and Drug, University of Parma, 43121 Parma, Italy
| | - Giovanni Zuliani
- Department of Translational Medicine and for Romagna, University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
23
|
Cochran BJ, Ong KL, Manandhar B, Rye KA. APOA1: a Protein with Multiple Therapeutic Functions. Curr Atheroscler Rep 2021; 23:11. [PMID: 33591433 DOI: 10.1007/s11883-021-00906-7] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/08/2021] [Indexed: 01/11/2023]
Abstract
PURPOSE OF THE REVIEW Apolipoprotein (APO) A1, the main apolipoprotein of plasma high-density lipoproteins (HDLs), has several well documented cardioprotective functions. A number of additional potentially beneficial functions of APOA1 have recently been identified. This review is concerned with the therapeutic potential of all of these functions in multiple disease states. RECENT FINDINGS Knowledge of the beneficial functions of APOA1 in atherosclerosis, thrombosis, diabetes, cancer, and neurological disorders is increasing exponentially. These insights have led to the development of clinically relevant peptides and APOA1-containing, synthetic reconstituted HDL (rHDL) preparations that mimic the functions of full-length APOA1. APOA1 is a multifunctional apolipoprotein that has therapeutic potential in several diseases. Translation of this knowledge into the clinic is likely to be dependent on the efficacy and bioavailability of small peptides and synthetic rHDL preparations that are currently under investigation, or in development.
Collapse
Affiliation(s)
- Blake J Cochran
- Lipid Research Group, School of Medical Sciences, Faculty of Medicine, University of New South Wales Sydney, Level 4E Wallace Wurth Building, Kensington, New South Wales, 2052, Australia
| | - Kwok-Leung Ong
- Lipid Research Group, School of Medical Sciences, Faculty of Medicine, University of New South Wales Sydney, Level 4E Wallace Wurth Building, Kensington, New South Wales, 2052, Australia
| | - Bikash Manandhar
- Lipid Research Group, School of Medical Sciences, Faculty of Medicine, University of New South Wales Sydney, Level 4E Wallace Wurth Building, Kensington, New South Wales, 2052, Australia
| | - Kerry-Anne Rye
- Lipid Research Group, School of Medical Sciences, Faculty of Medicine, University of New South Wales Sydney, Level 4E Wallace Wurth Building, Kensington, New South Wales, 2052, Australia.
| |
Collapse
|
24
|
Comparison of Plasma Lipoprotein Composition and Function in Cerebral Amyloid Angiopathy and Alzheimer's Disease. Biomedicines 2021; 9:biomedicines9010072. [PMID: 33445800 PMCID: PMC7828227 DOI: 10.3390/biomedicines9010072] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/31/2020] [Accepted: 01/08/2021] [Indexed: 12/12/2022] Open
Abstract
Cerebral amyloid angiopathy (CAA) refers to beta-amyloid (Aβ) deposition in brain vessels and is clinically the main cause of lobar intracerebral hemorrhage (ICH). Aβ can also accumulate in brain parenchyma forming neuritic plaques in Alzheimer’s disease (AD). Our study aimed to determine whether the peripheral lipid profile and lipoprotein composition are associated with cerebral beta-amyloidosis pathology and may reflect biological differences in AD and CAA. For this purpose, lipid and apolipoproteins levels were analyzed in plasma from 51 ICH-CAA patients (collected during the chronic phase of the disease), 60 AD patients, and 60 control subjects. Lipoproteins (VLDL, LDL, and HDL) were isolated and their composition and pro/antioxidant ability were determined. We observed that alterations in the lipid profile and lipoprotein composition were remarkable in the ICH-CAA group compared to control subjects, whereas the AD group presented no specific alterations compared with controls. ICH-CAA patients presented an atheroprotective profile, which consisted of lower total and LDL cholesterol levels. Plasma from chronic ICH-CAA patients also showed a redistribution of ApoC-III from HDL to VLDL and a higher ApoE/ApoC-III ratio in HDL. Whether these alterations reflect a protective response or have a causative effect on the pathology requires further investigation.
Collapse
|
25
|
Marsillach J, Adorni MP, Zimetti F, Papotti B, Zuliani G, Cervellati C. HDL Proteome and Alzheimer's Disease: Evidence of a Link. Antioxidants (Basel) 2020; 9:E1224. [PMID: 33287338 PMCID: PMC7761753 DOI: 10.3390/antiox9121224] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/25/2020] [Accepted: 11/30/2020] [Indexed: 12/16/2022] Open
Abstract
Several lines of epidemiological evidence link increased levels of high-density lipoprotein-cholesterol (HDL-C) with lower risk of Alzheimer's disease (AD). This observed relationship might reflect the beneficial effects of HDL on the cardiovascular system, likely due to the implication of vascular dysregulation in AD development. The atheroprotective properties of this lipoprotein are mostly due to its proteome. In particular, apolipoprotein (Apo) A-I, E, and J and the antioxidant accessory protein paraoxonase 1 (PON1), are the main determinants of the biological function of HDL. Intriguingly, these HDL constituent proteins are also present in the brain, either from in situ expression, or derived from the periphery. Growing preclinical evidence suggests that these HDL proteins may prevent the aberrant changes in the brain that characterize AD pathogenesis. In the present review, we summarize and critically examine the current state of knowledge on the role of these atheroprotective HDL-associated proteins in AD pathogenesis and physiopathology.
Collapse
Affiliation(s)
- Judit Marsillach
- Department of Environmental & Occupational Health Sciences, University of Washington, Seattle, WA 98195, USA;
| | - Maria Pia Adorni
- Unit of Neurosciences, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy;
| | - Francesca Zimetti
- Department of Food and Drug, University of Parma, 43124 Parma, Italy;
| | - Bianca Papotti
- Department of Food and Drug, University of Parma, 43124 Parma, Italy;
| | - Giovanni Zuliani
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (G.Z.); (C.C.)
| | - Carlo Cervellati
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (G.Z.); (C.C.)
| |
Collapse
|
26
|
Swaminathan SK, Zhou AL, Ahlschwede KM, Curran GL, Lowe VJ, Li L, Kandimalla KK. High-Density Lipoprotein Mimetic Peptide 4F Efficiently Crosses the Blood-Brain Barrier and Modulates Amyloid- β Distribution between Brain and Plasma. J Pharmacol Exp Ther 2020; 375:308-316. [PMID: 32778535 PMCID: PMC7589947 DOI: 10.1124/jpet.120.265876] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 07/24/2020] [Indexed: 12/24/2022] Open
Abstract
Treatments to elevate high-density lipoprotein (HDL) levels in plasma have decreased cerebrovascular amyloid -β (Aβ) deposition and mitigated cognitive decline in Alzheimer disease (AD) transgenic mice. Since the major protein component of HDL particles, apolipoprotein A-I (ApoA-I), has very low permeability at the blood-brain barrier (BBB), we investigated 4F, an 18-amino-acid ApoA-I/HDL mimetic peptide, as a therapeutic alternative. Specifically, we examined the BBB permeability of 4F and its effects on [125I]Aβ trafficking from brain to blood and from blood to brain. After systemic injection in mice, the BBB permeability of [125I]4F, estimated as the permeability-surface area (PS) product, ranged between 2 and 5 × 10-6 ml/g per second in various brain regions. The PS products of [125I]4F were ∼1000-fold higher compared with those determined for [125I]ApoA-I. Moreover, systemic infusion with 4F increased the brain efflux of intracerebrally injected [125I]Aβ42. Conversely, 4F infusion decreased the brain influx of systemically injected [125I]Aβ42. Interestingly, 4F did not significantly alter the brain influx of [125I]Aβ40. To corroborate the in vivo findings, we evaluated the effects of 4F on [125I]Aβ42 transcytosis across polarized human BBB endothelial cell (hCMEC/D3) monolayers. Treatment with 4F increased the abluminal-to-luminal flux and decreased the luminal-to-abluminal flux of [125I]Aβ42 across the hCMEC/D3 monolayers. Additionally, 4F decreased the endothelial accumulation of fluorescein-labeled Aβ42 in the hCMEC/D3 monolayers. These findings provide a mechanistic interpretation for the reductions in brain Aβ burden reported in AD mice after oral 4F administration, which represents a novel strategy for treating AD and cerebral amyloid angiopathy. SIGNIFICANCE STATEMENT: The brain permeability of the ApoA-I mimetic peptide 4F was estimated to be ∼1000-fold greater than ApoA-I after systemic injection of radiolabeled peptide/protein in mice. Further, 4F treatment increased the brain efflux of amyloid -β and also decreased its brain influx, as evaluated in mice and in blood-brain barrier cell monolayers. Thus, 4F represents a potential therapeutic strategy to mitigate brain amyloid accumulation in cerebral amyloid angiopathy and Alzheimer disease.
Collapse
Affiliation(s)
- Suresh K Swaminathan
- Department of Pharmaceutics and Brain Barriers Research Center (S.K.S., A.L.Z., K.M.A., K.K.K.) and Department of Experimental and Clinical Pharmacology (L.L.), University of Minnesota, College of Pharmacy, Minneapolis, Minnesota; Department of Pharmaceutical Sciences, College of Pharmacy, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois (K.M.A.); and Departments of Radiology (G.L.C., V.J.L.) and Neurology (G.L.C.), Mayo Clinic, College of Medicine, Rochester, Minnesota
| | - Andrew L Zhou
- Department of Pharmaceutics and Brain Barriers Research Center (S.K.S., A.L.Z., K.M.A., K.K.K.) and Department of Experimental and Clinical Pharmacology (L.L.), University of Minnesota, College of Pharmacy, Minneapolis, Minnesota; Department of Pharmaceutical Sciences, College of Pharmacy, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois (K.M.A.); and Departments of Radiology (G.L.C., V.J.L.) and Neurology (G.L.C.), Mayo Clinic, College of Medicine, Rochester, Minnesota
| | - Kristen M Ahlschwede
- Department of Pharmaceutics and Brain Barriers Research Center (S.K.S., A.L.Z., K.M.A., K.K.K.) and Department of Experimental and Clinical Pharmacology (L.L.), University of Minnesota, College of Pharmacy, Minneapolis, Minnesota; Department of Pharmaceutical Sciences, College of Pharmacy, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois (K.M.A.); and Departments of Radiology (G.L.C., V.J.L.) and Neurology (G.L.C.), Mayo Clinic, College of Medicine, Rochester, Minnesota
| | - Geoffry L Curran
- Department of Pharmaceutics and Brain Barriers Research Center (S.K.S., A.L.Z., K.M.A., K.K.K.) and Department of Experimental and Clinical Pharmacology (L.L.), University of Minnesota, College of Pharmacy, Minneapolis, Minnesota; Department of Pharmaceutical Sciences, College of Pharmacy, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois (K.M.A.); and Departments of Radiology (G.L.C., V.J.L.) and Neurology (G.L.C.), Mayo Clinic, College of Medicine, Rochester, Minnesota
| | - Val J Lowe
- Department of Pharmaceutics and Brain Barriers Research Center (S.K.S., A.L.Z., K.M.A., K.K.K.) and Department of Experimental and Clinical Pharmacology (L.L.), University of Minnesota, College of Pharmacy, Minneapolis, Minnesota; Department of Pharmaceutical Sciences, College of Pharmacy, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois (K.M.A.); and Departments of Radiology (G.L.C., V.J.L.) and Neurology (G.L.C.), Mayo Clinic, College of Medicine, Rochester, Minnesota
| | - Ling Li
- Department of Pharmaceutics and Brain Barriers Research Center (S.K.S., A.L.Z., K.M.A., K.K.K.) and Department of Experimental and Clinical Pharmacology (L.L.), University of Minnesota, College of Pharmacy, Minneapolis, Minnesota; Department of Pharmaceutical Sciences, College of Pharmacy, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois (K.M.A.); and Departments of Radiology (G.L.C., V.J.L.) and Neurology (G.L.C.), Mayo Clinic, College of Medicine, Rochester, Minnesota
| | - Karunya K Kandimalla
- Department of Pharmaceutics and Brain Barriers Research Center (S.K.S., A.L.Z., K.M.A., K.K.K.) and Department of Experimental and Clinical Pharmacology (L.L.), University of Minnesota, College of Pharmacy, Minneapolis, Minnesota; Department of Pharmaceutical Sciences, College of Pharmacy, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois (K.M.A.); and Departments of Radiology (G.L.C., V.J.L.) and Neurology (G.L.C.), Mayo Clinic, College of Medicine, Rochester, Minnesota
| |
Collapse
|
27
|
The Role of HDL and HDL Mimetic Peptides as Potential Therapeutics for Alzheimer's Disease. Biomolecules 2020; 10:biom10091276. [PMID: 32899606 PMCID: PMC7563116 DOI: 10.3390/biom10091276] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 08/25/2020] [Accepted: 08/31/2020] [Indexed: 12/11/2022] Open
Abstract
The role of high-density lipoproteins (HDL) in the cardiovascular system has been extensively studied and the cardioprotective effects of HDL are well established. As HDL particles are formed both in the systemic circulation and in the central nervous system, the role of HDL and its associated apolipoproteins in the brain has attracted much research interest in recent years. Alzheimer’s disease (AD) is the most prevalent neurodegenerative disorder and the leading cause of dementia worldwide, for which there currently exists no approved disease modifying treatment. Multiple lines of evidence, including a number of large-scale human clinical studies, have shown a robust connection between HDL levels and AD. Low levels of HDL are associated with increased risk and severity of AD, whereas high levels of HDL are correlated with superior cognitive function. Although the mechanisms underlying the protective effects of HDL in the brain are not fully understood, many of the functions of HDL, including reverse lipid/cholesterol transport, anti-inflammation/immune modulation, anti-oxidation, microvessel endothelial protection, and proteopathy modification, are thought to be critical for its beneficial effects. This review describes the current evidence for the role of HDL in AD and the potential of using small peptides mimicking HDL or its associated apolipoproteins (HDL-mimetic peptides) as therapeutics to treat AD.
Collapse
|
28
|
Valenzuela PL, Castillo-García A, Morales JS, de la Villa P, Hampel H, Emanuele E, Lista S, Lucia A. Exercise benefits on Alzheimer's disease: State-of-the-science. Ageing Res Rev 2020; 62:101108. [PMID: 32561386 DOI: 10.1016/j.arr.2020.101108] [Citation(s) in RCA: 188] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 05/29/2020] [Accepted: 06/06/2020] [Indexed: 01/15/2023]
Abstract
Although there is no unanimity, growing evidence supports the value of regular physical exercise to prevent Alzheimer's disease as well as cognitive decline in affected patients. Together with an introductory summary on epidemiological evidence, the aim of this review is to summarize the current knowledge on the potential biological mechanisms underlying exercise benefits in this condition. Regular physical exercise has proven to be beneficial for traditional cardiovascular risk factors (e.g., reduced vascular flow, diabetes) involved in the pathogenesis of Alzheimer's disease. Exercise also promotes neurogenesis via increases in exercise-induced metabolic factors (e.g., ketone bodies, lactate) and muscle-derived myokines (cathepsin-B, irisin), which in turn stimulate the production of neurotrophins such as brain-derived neurotrophic factor. Finally, regular exercise exerts anti-inflammatory effects and improves the brain redox status, thereby ameliorating the pathophysiological hallmarks of Alzheimer's disease (e.g., amyloid-β deposition). In summary, physical exercise might provide numerous benefits through different pathways that might, in turn, help prevent risk and progression of Alzheimer's disease. More evidence is needed, however, based on human studies.
Collapse
|
29
|
Chew H, Solomon VA, Fonteh AN. Involvement of Lipids in Alzheimer's Disease Pathology and Potential Therapies. Front Physiol 2020; 11:598. [PMID: 32581851 PMCID: PMC7296164 DOI: 10.3389/fphys.2020.00598] [Citation(s) in RCA: 189] [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: 02/28/2020] [Accepted: 05/14/2020] [Indexed: 12/15/2022] Open
Abstract
Lipids constitute the bulk of the dry mass of the brain and have been associated with healthy function as well as the most common pathological conditions of the brain. Demographic factors, genetics, and lifestyles are the major factors that influence lipid metabolism and are also the key components of lipid disruption in Alzheimer's disease (AD). Additionally, the most common genetic risk factor of AD, APOE ϵ4 genotype, is involved in lipid transport and metabolism. We propose that lipids are at the center of Alzheimer's disease pathology based on their involvement in the blood-brain barrier function, amyloid precursor protein (APP) processing, myelination, membrane remodeling, receptor signaling, inflammation, oxidation, and energy balance. Under healthy conditions, lipid homeostasis bestows a balanced cellular environment that enables the proper functioning of brain cells. However, under pathological conditions, dyshomeostasis of brain lipid composition can result in disturbed BBB, abnormal processing of APP, dysfunction in endocytosis/exocytosis/autophagocytosis, altered myelination, disturbed signaling, unbalanced energy metabolism, and enhanced inflammation. These lipid disturbances may contribute to abnormalities in brain function that are the hallmark of AD. The wide variance of lipid disturbances associated with brain function suggest that AD pathology may present as a complex interaction between several metabolic pathways that are augmented by risk factors such as age, genetics, and lifestyles. Herewith, we examine factors that influence brain lipid composition, review the association of lipids with all known facets of AD pathology, and offer pointers for potential therapies that target lipid pathways.
Collapse
Affiliation(s)
- Hannah Chew
- Huntington Medical Research Institutes, Pasadena, CA, United States
- University of California, Los Angeles, Los Angeles, CA, United States
| | | | - Alfred N. Fonteh
- Huntington Medical Research Institutes, Pasadena, CA, United States
| |
Collapse
|
30
|
Gisonno RA, Prieto ED, Gorgojo JP, Curto LM, Rodriguez ME, Rosú SA, Gaddi GM, Finarelli GS, Cortez MF, Schinella GR, Tricerri MA, Ramella NA. Fibrillar conformation of an apolipoprotein A-I variant involved in amyloidosis and atherosclerosis. Biochim Biophys Acta Gen Subj 2020; 1864:129515. [PMID: 31904503 DOI: 10.1016/j.bbagen.2020.129515] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 12/21/2019] [Accepted: 12/30/2019] [Indexed: 11/25/2022]
Abstract
BACKGROUND Different protein conformations may be involved in the development of clinical manifestations associated with human amyloidosis. Although a fibrillar conformation is usually the signature of damage in the tissues of patients, it is not clear whether this species is per se the cause or the consequence of the disease. Hereditary amyloidosis due to variants of apolipoprotein A-I (apoA-I) with a substitution of a single amino acid is characterized by the presence of fibrillar protein within the lesions. Thus mutations result in increased protein aggregation. Here we set up to characterize the folding of a natural variant with a mutation leading to a deletion at position 107 (apoA-I Lys107-0). Patients carrying this variant show amyloidosis and severe atherosclerosis. METHODS We oxidized this variant under controlled concentrations of hydrogen peroxide and analyzed the structure obtained after 30-day incubation by fluorescence, circular dichroism and microscopy approaches. Neutrophils activation was characterized by confocal microscopy. RESULTS We obtained a high yield of well-defined stable fibrillar structures of apoA-I Lys107-0. In an in vitro neutrophils system, we were able to detect the induction of Neutrophils Extracellular Traps (NETs) when we incubated with oxidized apoA-I variants. This effect was exacerbated by the fibrillar structure of oxidized Lys 107-0. CONCLUSIONS We conclude that a pro-inflammatory microenvironment could result in the formation of aggregation-prone species, which, in addition may induce a positive feed-back in the activation of an inflammatory response. GENERAL SIGNIFICANCE These events may explain a close association between amyloidosis due to apoA-I Lys107-0 and atherosclerosis.
Collapse
Affiliation(s)
- Romina A Gisonno
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), Argentina; Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Calle 60 y 120, La Plata, Argentina
| | - Eduardo D Prieto
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas (INIFTA), La Plata, Argentina
| | - Juan P Gorgojo
- Centro de Investigación y Desarrollo en Fermentaciones Industriales (CINDEFI), La Plata, Argentina
| | - Lucrecia M Curto
- Instituto de Química y Fisicoquímica Biológicas "Profesor Alejandro C. Paladini" (IQUIFIB) y Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, CABA, Argentina
| | - M Eugenia Rodriguez
- Centro de Investigación y Desarrollo en Fermentaciones Industriales (CINDEFI), La Plata, Argentina
| | - Silvana A Rosú
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), Argentina; Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Calle 60 y 120, La Plata, Argentina
| | - Gisela M Gaddi
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), Argentina; Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Calle 60 y 120, La Plata, Argentina
| | | | - M Fernanda Cortez
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), Argentina
| | - Guillermo R Schinella
- Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Calle 60 y 120, La Plata, Argentina
| | - M Alejandra Tricerri
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), Argentina; Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Calle 60 y 120, La Plata, Argentina.
| | - Nahuel A Ramella
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), Argentina; Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Calle 60 y 120, La Plata, Argentina.
| |
Collapse
|
31
|
Gao Q, Wang Y, Wang X, Fu S, Zhang X, Wang RT, Zhang X. Decreased levels of circulating trimethylamine N-oxide alleviate cognitive and pathological deterioration in transgenic mice: a potential therapeutic approach for Alzheimer's disease. Aging (Albany NY) 2019; 11:8642-8663. [PMID: 31612864 PMCID: PMC6814608 DOI: 10.18632/aging.102352] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 09/27/2019] [Indexed: 12/23/2022]
Abstract
Trimethylamine-N-oxide (TMAO), a metabolite of gut microbiota, has been implicated in the pathogenesis of Alzheimer’s disease (AD). However, the mechanisms by which TMAO influence cognitive and pathological processes in the AD have not been investigated. In this study, we found that the circulating TMAO levels displayed an age-related increase in both WT and APP/PS1 mice and association with AD-like behavioral and pathological profile. Reduced TMAO by 3,3-Dimethyl-1-butanol (DMB) treatment ameliorated the cognitive deterioration and long-term potentiation (LTP) in APP/PS1 mice. Moreover, DMB treatment also induced a decrease in the Amyloid-β (Aβ)1-42, β-secretase, and β-secretase-cleaved C-terminal fragment (βCTF) levels in the hippocampus. Finally, the effects obtained after treatment with DMB were accompanied by a reduction in circulating clusterin levels and hippocampal neuroinflammatory status in APP/PS1 mice. These findings demonstrate that elevated circulating TMAO during the aging process might deteriorate cognitive function and pathology in APP/PS1 mice.
Collapse
Affiliation(s)
- Qiang Gao
- Department of Geriatrics, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Yuan Wang
- Department of Geriatrics, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Xin Wang
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin 150081, China
| | - Shuang Fu
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin 150081, China
| | - Xin Zhang
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin 150081, China
| | - Rui-Tao Wang
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin 150081, China
| | - Xin Zhang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| |
Collapse
|
32
|
Brain-targeted drug delivery by manipulating protein corona functions. Nat Commun 2019; 10:3561. [PMID: 31395892 PMCID: PMC6687821 DOI: 10.1038/s41467-019-11593-z] [Citation(s) in RCA: 187] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 07/23/2019] [Indexed: 12/21/2022] Open
Abstract
Protein corona presents a major obstacle to bench-to-bedside translation of targeted drug delivery systems, severely affecting targeting yields and directing unfavorable biodistribution. Corona-mediated targeting provides a new impetus for specific drug delivery by precisely manipulating interaction modes of functional plasma proteins on nano-surface. Here bio-inspired liposomes (SP-sLip) were developed by modifying liposomal surface with a short nontoxic peptide derived from Aβ1-42 that specifically interacts with the lipid-binding domain of exchangeable apolipoproteins. SP-sLip absorb plasma apolipoproteins A1, E and J, consequently exposing receptor-binding domain of apolipoproteins to achieve brain-targeted delivery. Doxorubicin loaded SP-sLip (SP-sLip/DOX) show significant enhancement of brain distribution and anti-brain cancer effect in comparison to doxorubicin loaded plain liposomes. SP-sLip preserve functions of the absorbed human plasma ApoE, and the corona-mediated targeting strategy works in SP modified PLGA nanoparticles. The present study may pave a new avenue to facilitate clinical translation of targeted drug delivery systems. Plasma proteins may severely affect the in vivo performance of liposomes. Here, the authors develop bio-inspired liposomes that specifically absorb brain-targeted apolipoproteins and preserve their bioactivities, thereby achieving efficient brain targeting with minor influence on immunocompatibility of liposomes.
Collapse
|
33
|
Slot RE, Kester MI, Van Harten AC, Jongbloed W, Bouwman FH, Teunissen CE, Scheltens P, van der Flier WM, Veerhuis R. ApoE and clusterin CSF levels influence associations between APOE genotype and changes in CSF tau, but not CSF Aβ42, levels in non-demented elderly. Neurobiol Aging 2019; 79:101-109. [DOI: 10.1016/j.neurobiolaging.2019.02.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 02/18/2019] [Accepted: 02/22/2019] [Indexed: 01/14/2023]
|
34
|
Zhou AL, Swaminathan SK, Curran GL, Poduslo JF, Lowe VJ, Li L, Kandimalla KK. Apolipoprotein A-I Crosses the Blood-Brain Barrier through Clathrin-Independent and Cholesterol-Mediated Endocytosis. J Pharmacol Exp Ther 2019; 369:481-488. [PMID: 30971477 PMCID: PMC6538888 DOI: 10.1124/jpet.118.254201] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 03/11/2019] [Indexed: 11/22/2022] Open
Abstract
Recent studies suggest that apolipoprotein A-I (ApoA-I), the major protein constituent of high-density lipoprotein particles, plays a critical role in preserving cerebrovascular integrity and reducing Alzheimer's risk. ApoA-I present in brain is thought to be primarily derived from the peripheral circulation. Although plasma-to-brain delivery of ApoA-I is claimed to be handled by the blood-cerebrospinal fluid barrier (BCSFB), a contribution by the blood-brain barrier (BBB), which serves as a major portal for protein delivery to brain, cannot be ruled out. In this study, we assessed the permeability-surface area product (PS) of radioiodinated ApoA-I (125I-ApoA-I) in various brain regions of wild-type rats after an intravenous bolus injection. The PS value at the cortex, caudate putamen, hippocampus, thalamus, brain stem, and cerebellum was found to be 0.39, 0.28, 0.28, 0.36, 0.69, and 0.76 (ml/g per second × 10-6), respectively. Solutes delivered into brain via the BCSFB are expected to show greater accumulation in the thalamus due to its periventricular location. The modest permeability for 125I-ApoA-I into the thalamus relative to other regions suggests that BCSFB transport accounts for only a portion of total brain uptake and thus BBB transport cannot be ruled out. In addition, we show that Alexa Flour 647-labeled ApoA-I (AF647-ApoA-I) undergoes clathrin-independent and cholesterol-mediated endocytosis in transformed human cerebral microvascular endothelial cells (hCMEC/D3). Further, Z-series confocal images of the hCMEC/D3 monolayers and Western blot detection of intact ApoA-I on the abluminal side demonstrated AF647-ApoA-I transcytosis across the endothelium. These findings implicate the BBB as a significant portal for ApoA-I delivery into brain.
Collapse
Affiliation(s)
- Andrew L Zhou
- Department of Pharmaceutics and Brain Barriers Research Center (A.L.Z., S.K.S., K.K.K.) and Department of Experimental and Clinical Pharmacology (L.L.), College of Pharmacy, University of Minnesota, Minneapolis, Minnesota; and Department of Radiology (G.L.C., V.J.L.) and Department of Neurology (G.L.C., J.F.P.), Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Suresh K Swaminathan
- Department of Pharmaceutics and Brain Barriers Research Center (A.L.Z., S.K.S., K.K.K.) and Department of Experimental and Clinical Pharmacology (L.L.), College of Pharmacy, University of Minnesota, Minneapolis, Minnesota; and Department of Radiology (G.L.C., V.J.L.) and Department of Neurology (G.L.C., J.F.P.), Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Geoffry L Curran
- Department of Pharmaceutics and Brain Barriers Research Center (A.L.Z., S.K.S., K.K.K.) and Department of Experimental and Clinical Pharmacology (L.L.), College of Pharmacy, University of Minnesota, Minneapolis, Minnesota; and Department of Radiology (G.L.C., V.J.L.) and Department of Neurology (G.L.C., J.F.P.), Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Joseph F Poduslo
- Department of Pharmaceutics and Brain Barriers Research Center (A.L.Z., S.K.S., K.K.K.) and Department of Experimental and Clinical Pharmacology (L.L.), College of Pharmacy, University of Minnesota, Minneapolis, Minnesota; and Department of Radiology (G.L.C., V.J.L.) and Department of Neurology (G.L.C., J.F.P.), Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Val J Lowe
- Department of Pharmaceutics and Brain Barriers Research Center (A.L.Z., S.K.S., K.K.K.) and Department of Experimental and Clinical Pharmacology (L.L.), College of Pharmacy, University of Minnesota, Minneapolis, Minnesota; and Department of Radiology (G.L.C., V.J.L.) and Department of Neurology (G.L.C., J.F.P.), Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Ling Li
- Department of Pharmaceutics and Brain Barriers Research Center (A.L.Z., S.K.S., K.K.K.) and Department of Experimental and Clinical Pharmacology (L.L.), College of Pharmacy, University of Minnesota, Minneapolis, Minnesota; and Department of Radiology (G.L.C., V.J.L.) and Department of Neurology (G.L.C., J.F.P.), Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Karunya K Kandimalla
- Department of Pharmaceutics and Brain Barriers Research Center (A.L.Z., S.K.S., K.K.K.) and Department of Experimental and Clinical Pharmacology (L.L.), College of Pharmacy, University of Minnesota, Minneapolis, Minnesota; and Department of Radiology (G.L.C., V.J.L.) and Department of Neurology (G.L.C., J.F.P.), Mayo Clinic College of Medicine, Rochester, Minnesota
| |
Collapse
|
35
|
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
|
36
|
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
|
37
|
de Retana SF, Marazuela P, Solé M, Colell G, Bonaterra A, Sánchez-Quesada JL, Montaner J, Maspoch D, Cano-Sarabia M, Hernández-Guillamon M. Peripheral administration of human recombinant ApoJ/clusterin modulates brain beta-amyloid levels in APP23 mice. ALZHEIMERS RESEARCH & THERAPY 2019; 11:42. [PMID: 31077261 PMCID: PMC6511153 DOI: 10.1186/s13195-019-0498-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 04/23/2019] [Indexed: 12/15/2022]
Abstract
Background ApoJ/clusterin is a multifunctional protein highly expressed in the brain. The implication of ApoJ in β-amyloid (Aβ) fibrillization and clearance in the context of Alzheimer’s disease has been widely studied, although the source and concentration of ApoJ that promotes or inhibits Aβ cerebral accumulation is not clear yet. ApoJ is abundant in plasma and approximately 20% can appear bound to HDL-particles. In this regard, the impact of plasmatic ApoJ and its lipidation status on cerebral β-amyloidosis is still not known. Hence, our main objective was to study the effect of a peripheral increase of free ApoJ or reconstituted HDL particles containing ApoJ in an experimental model of cerebral β-amyloidosis. Methods Fourteen-month-old APP23 transgenic mice were subjected to subchronic intravenous treatment with rHDL-rApoJ nanodiscs or free rApoJ for 1 month. Aβ concentration and distribution in the brain, as well as Aβ levels in plasma and CSF, were determined after treatments. Other features associated to AD pathology, such as neuronal loss and neuroinflammation, were also evaluated. Results Both ApoJ-based treatments prevented the Aβ accumulation in cerebral arteries and induced a decrease in total brain insoluble Aβ42 levels. The peripheral treatment with rApoJ also induced an increase in the Aβ40 levels in CSF, whereas the concentration remained unaltered in plasma. At all the endpoints studied, the lipidation of rApoJ did not enhance the protective properties of free rApoJ. The effects obtained after subchronic treatment with free rApoJ were accompanied by a reduction in hippocampal neuronal loss and an enhancement of the expression of a phagocytic marker in microglial cells surrounding Aβ deposits. Finally, despite the activation of this phagocytic phenotype, treatments did not induce a global neuroinflammatory status. In fact, free rApoJ treatment was able to reduce the levels of interleukin-17 (IL17) and keratinocyte chemoattractant (KC) chemokine in the brain. Conclusions Our results demonstrate that an increase in circulating human rApoJ induces a reduction of insoluble Aβ and CAA load in the brain of APP23 mice. Thus, our study suggests that peripheral interventions, based on treatments with multifunctional physiological chaperones, offer therapeutic opportunities to regulate the cerebral Aβ load. Electronic supplementary material The online version of this article (10.1186/s13195-019-0498-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sofía Fernández de Retana
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, 08035, Barcelona, Spain
| | - Paula Marazuela
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, 08035, Barcelona, Spain
| | - Montse Solé
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, 08035, Barcelona, Spain
| | - Guillem Colell
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, 08035, Barcelona, Spain
| | - Anna Bonaterra
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, 08035, Barcelona, Spain
| | - Jose Luis Sánchez-Quesada
- Cardiovascular Biochemistry Group, Research Institute of the Hospital de Sant Pau (IIB Sant Pau), Barcelona, Spain.,CIBER of Diabetes and Metabolism (CIBERDEM), ISCIII, Madrid, Spain
| | - Joan Montaner
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, 08035, Barcelona, Spain
| | - Daniel Maspoch
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and the Barcelona Institute of Science and Technology, , Campus UAB, Bellaterra, Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), 08100, Barcelona, Spain
| | - Mary Cano-Sarabia
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and the Barcelona Institute of Science and Technology, , Campus UAB, Bellaterra, Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), 08100, Barcelona, Spain
| | - Mar Hernández-Guillamon
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, 08035, Barcelona, Spain.
| |
Collapse
|
38
|
Camacho J, Moliné T, Bonaterra-Pastra A, Ramón Y Cajal S, Martínez-Sáez E, Hernández-Guillamon M. Brain ApoA-I, ApoJ and ApoE Immunodetection in Cerebral Amyloid Angiopathy. Front Neurol 2019; 10:187. [PMID: 30918495 PMCID: PMC6424885 DOI: 10.3389/fneur.2019.00187] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 02/14/2019] [Indexed: 11/13/2022] Open
Abstract
Cerebral amyloid angiopathy (CAA) is a common cause of lobar intracerebral hemorrhage (ICH) in elderly individuals and it is the result of the cerebrovascular deposition of beta-amyloid (Aβ) protein. CAA is frequently found in patients with Alzheimer's disease (AD), although it has an independent contribution to the cognitive deterioration associated with age. Specific apolipoproteins (Apo) have been associated with Aβ fibrillization and clearance from the brain. In this regard, in the present study, we analyzed the brain levels of ApoE, ApoA-I, and ApoJ/clusterin in autopsy brains from 20 post-mortem cases with CAA type I, CAA type II, with parenchymal Aβ deposits or without Aβ deposits. Our objective was to find a possible differential pattern of apolipoproteins distribution in the brain depending on the CAA pathological presentation. The protein expression levels were adjusted by the APOE genotype of the patients included in the study. We found that ApoE and ApoJ were abundantly present in meningeal, cortical, and capillary vessels of the brains with vascular Aβ accumulation. ApoE and ApoJ also deposited extracellularly in the parenchyma, especially in cases presenting Aβ diffuse and neuritic parenchymal deposits. In contrast, ApoA-I staining was only relevant in capillary walls in CAA type I cases. On the other hand, ICH was the principal cause of death among CAA patients in our cohort. We found that CAA patients with ICH more commonly had APOEε2 compared with CAA patients without ICH. In addition, patients who suffered an ICH presented higher vascular ApoE levels in brain. However, higher ApoE presence in cortical arteries was the only independent predictor of suffering an ICH in our cohort after adjusting by age and APOE genotype. In conclusion, while ApoE and ApoJ appear to be involved in both vascular and parenchymal Aβ pathology, ApoA-I seems to be mainly associated with CAA, especially in CAA type I pathology. We consider that our study helps to molecularly characterize the distribution subtypes of Aβ deposition within the brain.
Collapse
Affiliation(s)
- Jessica Camacho
- Pathology Department, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Teresa Moliné
- Pathology Department, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Anna Bonaterra-Pastra
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Santiago Ramón Y Cajal
- Pathology Department, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Elena Martínez-Sáez
- Pathology Department, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Mar Hernández-Guillamon
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
39
|
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: 233] [Impact Index Per Article: 38.8] [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
|
40
|
Lamartinière Y, Boucau MC, Dehouck L, Krohn M, Pahnke J, Candela P, Gosselet F, Fenart L. ABCA7 Downregulation Modifies Cellular Cholesterol Homeostasis and Decreases Amyloid-β Peptide Efflux in an in vitro Model of the Blood-Brain Barrier. J Alzheimers Dis 2018; 64:1195-1211. [DOI: 10.3233/jad-170883] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Yordenca Lamartinière
- Université d’Artois, EA 2465, Laboratoire de la Barrière Hémato-Encéphalique (LBHE), France
| | - Marie-Christine Boucau
- Université d’Artois, EA 2465, Laboratoire de la Barrière Hémato-Encéphalique (LBHE), France
| | - Lucie Dehouck
- Université d’Artois, EA 2465, Laboratoire de la Barrière Hémato-Encéphalique (LBHE), France
| | - Markus Krohn
- Department of Neuro-/Pathology, University of Oslo (UiO) & Oslo University Hospital (OUS), Oslo, Norway
| | - Jens Pahnke
- Department of Neuro-/Pathology, University of Oslo (UiO) & Oslo University Hospital (OUS), Oslo, Norway
- University of Lübeck (UzL), LIED, Lübeck, Germany
- Leibniz Institute of Plant Biochemistry (IPB), Halle, Germany
| | - Pietra Candela
- Université d’Artois, EA 2465, Laboratoire de la Barrière Hémato-Encéphalique (LBHE), France
| | - Fabien Gosselet
- Université d’Artois, EA 2465, Laboratoire de la Barrière Hémato-Encéphalique (LBHE), France
| | - Laurence Fenart
- Université d’Artois, EA 2465, Laboratoire de la Barrière Hémato-Encéphalique (LBHE), France
| |
Collapse
|
41
|
Zandl-Lang M, Fanaee-Danesh E, Sun Y, Albrecher NM, Gali CC, Čančar I, Kober A, Tam-Amersdorfer C, Stracke A, Storck SM, Saeed A, Stefulj J, Pietrzik CU, Wilson MR, Björkhem I, Panzenboeck U. Regulatory effects of simvastatin and apoJ on APP processing and amyloid-β clearance in blood-brain barrier endothelial cells. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1863:40-60. [DOI: 10.1016/j.bbalip.2017.09.008] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 08/31/2017] [Accepted: 09/18/2017] [Indexed: 10/18/2022]
|
42
|
Fernández-de-Retana S, Cano-Sarabia M, Marazuela P, Sánchez-Quesada JL, Garcia-Leon A, Montañola A, Montaner J, Maspoch D, Hernández-Guillamon M. Characterization of ApoJ-reconstituted high-density lipoprotein (rHDL) nanodisc for the potential treatment of cerebral β-amyloidosis. Sci Rep 2017; 7:14637. [PMID: 29116115 PMCID: PMC5677083 DOI: 10.1038/s41598-017-15215-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 10/24/2017] [Indexed: 01/12/2023] Open
Abstract
Cerebral β-amyloidosis is a major feature of Alzheimer’s disease (AD), characterized by the accumulation of β-amyloid protein (Aβ) in the brain. Several studies have implicated lipid/lipoprotein metabolism in the regulation of β-amyloidosis. In this regard, HDL (High Density Lipoprotein)-based therapies could ameliorate pathological features associated with AD. As apolipoprotein J (ApoJ) is a natural chaperone that interacts with Aβ, avoiding its aggregation and toxicity, in this study we propose to prepare reconstituted rHDL-rApoJ nanoparticles by assembling phospholipids with recombinant human ApoJ (rApoJ). Hence, rHDL particles were prepared using the cholate dialysis method and characterized by N-PAGE, dynamic light scattering, circular dichroism and electron transmission microscopy. The preparation of rHDL particles showed two-sized populations with discoidal shape. Functionally, rHDL-rApoJ maintained the ability to prevent the Aβ fibrillization and mediated a higher cholesterol efflux from cultured macrophages. Fluorescently-labelled rHDL-rApoJ nanoparticles were intravenously administrated in mice and their distribution over time was determined using an IVIS Xenogen® imager. It was confirmed that rHDL-rApoJ accumulated in the cranial region, especially in old transgenic mice presenting a high cerebral Aβ load. In conclusion, we have standardized a reproducible protocol to produce rHDL-rApoJ nanoparticles, which may be potentially considered as a therapeutic option for β-amyloid-related pathologies.
Collapse
Affiliation(s)
- Sofía Fernández-de-Retana
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Mary Cano-Sarabia
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and the Barcelona Institute of Science and Technology, Campus UAB, Bellaterra, Barcelona, Spain.
| | - Paula Marazuela
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Jose Luis Sánchez-Quesada
- Cardiovascular Biochemistry Group, Research Institute of the Hospital de Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - Annabel Garcia-Leon
- Cardiovascular Biochemistry Group, Research Institute of the Hospital de Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - Alex Montañola
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Joan Montaner
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Daniel Maspoch
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and the Barcelona Institute of Science and Technology, Campus UAB, Bellaterra, Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), 08100, Barcelona, Spain
| | - Mar Hernández-Guillamon
- Neurovascular Research Laboratory, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain.
| |
Collapse
|
43
|
Fernández-de Retana S, Montañola A, Marazuela P, De La Cuesta M, Batlle A, Fatar M, Grudzenski S, Montaner J, Hernández-Guillamon M. Intravenous treatment with human recombinant ApoA-I Milano reduces beta amyloid cerebral deposition in the APP23-transgenic mouse model of Alzheimer's disease. Neurobiol Aging 2017; 60:116-128. [PMID: 28941727 DOI: 10.1016/j.neurobiolaging.2017.08.028] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 08/18/2017] [Accepted: 08/26/2017] [Indexed: 12/31/2022]
Abstract
Beyond the crucial role of apolipoprotein A-I (ApoA-I) on peripheral cholesterol metabolism, this apolipoprotein has also been implicated in beta amyloid (Aβ)-related neuropathologies. ApoA-I-Milano (M) is a mutated variant, which showed increased vasoprotective properties compared to ApoA-I-wild type in models of atherosclerosis and cardiovascular damage. We speculated that ApoA-I-M may also protect Aβ-affected vasculature and reverse some of the pathological features associated with Alzheimer's disease (AD). For this purpose, we produced and characterized human recombinant ApoA-I-wild type and ApoA-I-M proteins. Both of them were able to avoid the aggregation of Aβ in vitro, even though recombinant ApoA-I-M was significantly more effective in protecting endothelial cells from Aβ(1-42)-toxicity. Next, we determined the effect of chronic intravenous administration of rApoA-I-M in the APP23-transgenic mouse model of AD. We found reduced cerebral Aβ levels in mice that received rApoA-I-M, which were accompanied by a lower expression of astrocyte and microglia neuroinflammatory markers. Our results suggest an applicability of this molecule as a therapeutic candidate for protecting the brain in AD.
Collapse
Affiliation(s)
- Sofía Fernández-de Retana
- Neurovascular Research Laboratory, Vall d'Hebron Research Insitute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alex Montañola
- Neurovascular Research Laboratory, Vall d'Hebron Research Insitute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Paula Marazuela
- Neurovascular Research Laboratory, Vall d'Hebron Research Insitute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Maialen De La Cuesta
- Neurovascular Research Laboratory, Vall d'Hebron Research Insitute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Aina Batlle
- Neurovascular Research Laboratory, Vall d'Hebron Research Insitute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Marc Fatar
- Department of Neurology, Universitätsmedizin Mannheim, Heidelberg University, Mannheim, Germany
| | - Saskia Grudzenski
- Department of Neurology, Universitätsmedizin Mannheim, Heidelberg University, Mannheim, Germany
| | - Joan Montaner
- Neurovascular Research Laboratory, Vall d'Hebron Research Insitute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Mar Hernández-Guillamon
- Neurovascular Research Laboratory, Vall d'Hebron Research Insitute, Universitat Autònoma de Barcelona, Barcelona, Spain.
| |
Collapse
|
44
|
Qi XM, Ma JF. The role of amyloid beta clearance in cerebral amyloid angiopathy: more potential therapeutic targets. Transl Neurodegener 2017; 6:22. [PMID: 28824801 PMCID: PMC5559841 DOI: 10.1186/s40035-017-0091-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 08/01/2017] [Indexed: 01/09/2023] Open
Abstract
Cerebral amyloid angiopathy (CAA) is characterized by the deposition of amyloid β-protein (Aβ) in the leptomeningeal and cortical blood vessels, which is an age-dependent risk factor for intracerebral hemorrhage (ICH), ischemic stroke and contributes to cerebrovascular dysfunction leading to cognitive impairment. However clinical prevention and treatment of the disease is very difficult because of its occult onset and severity of the symptoms. In recent years, many anti-amyloid β immunotherapies have not demonstrated clinical efficacy in subjects with Alzheimer’s disease (AD), and the failure may be due to the deposition of Aβ in the cerebrovascular export pathway resulting in further damage to blood vessels and aggravating CAA. So decreased clearance of Aβ in blood vessels plays a crucial role in the development of CAA and AD, and identification of the molecular pathways involved will provide new targets for treatment. In this review, we mainly describe the mechanisms of Aβ clearance through vessels, especially in terms of some proteins and receptors involved in this process.
Collapse
Affiliation(s)
- Xue-Mei Qi
- Department of Neurology & Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China
| | - Jian-Fang Ma
- Department of Neurology & Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China
| |
Collapse
|
45
|
Boyce G, Button E, Soo S, Wellington C. The pleiotropic vasoprotective functions of high density lipoproteins (HDL). J Biomed Res 2017; 32:164. [PMID: 28550271 PMCID: PMC6265396 DOI: 10.7555/jbr.31.20160103] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 12/23/2016] [Indexed: 12/19/2022] Open
Abstract
The pleiotropic functions of circulating high density lipoprotein (HDL) on peripheral vascular health are well established. HDL plays a pivotal role in reverse cholesterol transport and is also known to suppress inflammation, endothelial activation and apoptosis in peripheral vessels. Although not expressed in the central nervous system, HDL has nevertheless emerged as a potential resilience factor for dementia in multiple epidemiological studies. Animal model data specifically support a role for HDL in attenuating the accumulation of β-amyloid within cerebral vessels concomitant with reduced neuroinflammation and improved cognitive performance. As the vascular contributions to dementia are increasingly appreciated, this review seeks to summarize recent literature focused on the vasoprotective properties of HDL that may extend to cerebral vessels, discuss potential roles of HDL in dementia relative to brain-derived lipoproteins, identify gaps in current knowledge, and highlight new opportunities for research and discovery.
Collapse
Affiliation(s)
- Guilaine Boyce
- Department of Pathology and Laboratory Medicine, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Emily Button
- Department of Pathology and Laboratory Medicine, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Sonja Soo
- Department of Pathology and Laboratory Medicine, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Cheryl Wellington
- Department of Pathology and Laboratory Medicine, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| |
Collapse
|
46
|
Kane JP, Malloy MJ. Lipoproteins and amyloid vascular disease. Curr Opin Lipidol 2016; 27:640-641. [PMID: 27805977 DOI: 10.1097/mol.0000000000000364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- John P Kane
- Cardiovascular Research Institute; University of California Medical Center, San Francisco, California, USA
| | | |
Collapse
|
47
|
Biemans EALM, Jäkel L, de Waal RMW, Kuiperij HB, Verbeek MM. Limitations of the hCMEC/D3 cell line as a model for Aβ clearance by the human blood-brain barrier. J Neurosci Res 2016; 95:1513-1522. [PMID: 27726164 PMCID: PMC5484315 DOI: 10.1002/jnr.23964] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 09/19/2016] [Accepted: 09/19/2016] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease and cerebral amyloid angiopathy are characterized by accumulation of amyloid-β (Aβ) at the cerebrovasculature due to decreased clearance at the blood-brain barrier (BBB). However, the exact mechanism of Aβ clearance across this barrier has not been fully elucidated. The hCMEC/D3 cell line has been characterized as a valid model for the BBB. In this study we evaluated the use of this model to study Aβ clearance across the BBB, with an emphasis on brain-to-blood directional permeability. Barrier integrity of hCMEC/D3 monolayers was confirmed for large molecules in both the apical to basolateral and the reverse direction. However, permeability for smaller molecules was substantially higher, especially in basolateral to apical direction, and barrier formation for Aβ was completely absent in this direction. In addition, hCMEC/D3 cells failed to develop a high TEER, possibly caused by incomplete formation of tight junctions. We conclude that the hCMEC/D3 model has several limitations to study the cerebral clearance of Aβ. Therefore, the model needs further characterization before this cell system can be generally applied as a model to study cerebral Aβ clearance. © 2016 The Authors Journal of Neuroscience Research Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Elisanne A L M Biemans
- Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Departments of Neurology and Laboratory Medicine, Radboud Alzheimer Centre, Nijmegen, The Netherlands
| | - Lieke Jäkel
- Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Departments of Neurology and Laboratory Medicine, Radboud Alzheimer Centre, Nijmegen, The Netherlands
| | - Robert M W de Waal
- Radboud University Medical Center, Department of Pathology, Nijmegen, The Netherlands
| | - H Bea Kuiperij
- Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Departments of Neurology and Laboratory Medicine, Radboud Alzheimer Centre, Nijmegen, The Netherlands
| | - Marcel M Verbeek
- Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Departments of Neurology and Laboratory Medicine, Radboud Alzheimer Centre, Nijmegen, The Netherlands
| |
Collapse
|