1
|
Wu B, Liu Y, Li H, Zhu L, Zeng L, Zhang Z, Peng W. Liver as a new target organ in Alzheimer's disease: insight from cholesterol metabolism and its role in amyloid-beta clearance. Neural Regen Res 2025; 20:695-714. [PMID: 38886936 PMCID: PMC11433892 DOI: 10.4103/1673-5374.391305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 11/07/2023] [Indexed: 06/20/2024] Open
Abstract
Alzheimer's disease, the primary cause of dementia, is characterized by neuropathologies, such as amyloid plaques, synaptic and neuronal degeneration, and neurofibrillary tangles. Although amyloid plaques are the primary characteristic of Alzheimer's disease in the central nervous system and peripheral organs, targeting amyloid-beta clearance in the central nervous system has shown limited clinical efficacy in Alzheimer's disease treatment. Metabolic abnormalities are commonly observed in patients with Alzheimer's disease. The liver is the primary peripheral organ involved in amyloid-beta metabolism, playing a crucial role in the pathophysiology of Alzheimer's disease. Notably, impaired cholesterol metabolism in the liver may exacerbate the development of Alzheimer's disease. In this review, we explore the underlying causes of Alzheimer's disease and elucidate the role of the liver in amyloid-beta clearance and cholesterol metabolism. Furthermore, we propose that restoring normal cholesterol metabolism in the liver could represent a promising therapeutic strategy for addressing Alzheimer's disease.
Collapse
Affiliation(s)
- Beibei Wu
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Yuqing Liu
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Hongli Li
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Lemei Zhu
- Academician Workstation, Changsha Medical University, Changsha, Hunan Province, China
| | - Lingfeng Zeng
- Academician Workstation, Changsha Medical University, Changsha, Hunan Province, China
| | - Zhen Zhang
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Yangsheng College of Traditional Chinese Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou Province, China
- Qinhuangdao Shanhaiguan Pharmaceutical Co., Ltd, Qinhuangdao, Hebei Province, China
| | - Weijun Peng
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- National Clinical Research Center for Mental Disorder, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| |
Collapse
|
2
|
Velma GR, Laham MS, Lewandowski C, Valencia-Olvera AC, Balu D, Moore A, Ackerman-Berrier M, Rychetsky P, Penton C, Musku SR, Annadurai A, Sulaiman MI, Ma N, J Thatcher GR. Nonlipogenic ABCA1 Inducers (NLAI) for Alzheimer's Disease Validated in a Mouse Model Expressing Human APOE3/APOE4. J Med Chem 2024; 67:15061-15079. [PMID: 39191400 PMCID: PMC11404489 DOI: 10.1021/acs.jmedchem.4c00733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
Therapeutics enhancing apolipoprotein (APOE) positive function are a priority, because APOE4 is the major genetic risk factor for Alzheimer's disease (AD). The function of APOE, the key constituent of lipoprotein particles that transport cholesterol and lipids in the brain, is dependent on lipidation by ABCA1, a cell-membrane cholesterol transporter. ABCA1 transcription is regulated by liver X receptors (LXR): agonists have been shown to increase ABCA1, often accompanied by unwanted lipogenesis and elevated triglycerides (TG). Therefore, nonlipogenic ABCA1-inducers (NLAI) are needed. Two rounds of optimization of an HTS hit, derived from a phenotypic screen, gave lead compound 39 that was validated and tested in E3/4FAD mice that express human APOE3/4 and five mutant APP and PSEN1 human transgenes. Treatment with 39 increased ABCA1 expression, enhanced APOE lipidation, and reversed multiple AD phenotypes, without increasing TG. This NLAI/LXR-agonist study is the first in a human APOE-expressing model with hallmark amyloid-β pathology.
Collapse
Affiliation(s)
- Ganga Reddy Velma
- Department of Pharmacology & Toxicology, R Ken Coit College of Pharmacy, University of Arizona, Tucson, Arizona 85721, United States
| | - Megan S Laham
- Department of Chemistry & Biochemistry, Colleges of Science & Medicine, University of Arizona, Tucson, Arizona 85721, United States
| | - Cutler Lewandowski
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Ana C Valencia-Olvera
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Deebika Balu
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Annabelle Moore
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Martha Ackerman-Berrier
- Department of Pharmacology & Toxicology, R Ken Coit College of Pharmacy, University of Arizona, Tucson, Arizona 85721, United States
| | - Pavel Rychetsky
- Department of Pharmacology & Toxicology, R Ken Coit College of Pharmacy, University of Arizona, Tucson, Arizona 85721, United States
| | - Christopher Penton
- Department of Pharmacology & Toxicology, R Ken Coit College of Pharmacy, University of Arizona, Tucson, Arizona 85721, United States
| | - Soumya Reddy Musku
- Department of Pharmacology & Toxicology, R Ken Coit College of Pharmacy, University of Arizona, Tucson, Arizona 85721, United States
| | - Anandhan Annadurai
- Department of Pharmacology & Toxicology, R Ken Coit College of Pharmacy, University of Arizona, Tucson, Arizona 85721, United States
| | - Maha Ibrahim Sulaiman
- Department of Pharmacology & Toxicology, R Ken Coit College of Pharmacy, University of Arizona, Tucson, Arizona 85721, United States
| | - Nina Ma
- Department of Pharmacology & Toxicology, R Ken Coit College of Pharmacy, University of Arizona, Tucson, Arizona 85721, United States
| | - Gregory R J Thatcher
- Department of Pharmacology & Toxicology, R Ken Coit College of Pharmacy, University of Arizona, Tucson, Arizona 85721, United States
- Department of Chemistry & Biochemistry, Colleges of Science & Medicine, University of Arizona, Tucson, Arizona 85721, United States
| |
Collapse
|
3
|
Loeffler DA. Enhancing of cerebral Abeta clearance by modulation of ABC transporter expression: a review of experimental approaches. Front Aging Neurosci 2024; 16:1368200. [PMID: 38872626 PMCID: PMC11170721 DOI: 10.3389/fnagi.2024.1368200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 05/01/2024] [Indexed: 06/15/2024] Open
Abstract
Clearance of amyloid-beta (Aβ) from the brain is impaired in both early-onset and late-onset Alzheimer's disease (AD). Mechanisms for clearing cerebral Aβ include proteolytic degradation, antibody-mediated clearance, blood brain barrier and blood cerebrospinal fluid barrier efflux, glymphatic drainage, and perivascular drainage. ATP-binding cassette (ABC) transporters are membrane efflux pumps driven by ATP hydrolysis. Their functions include maintenance of brain homeostasis by removing toxic peptides and compounds, and transport of bioactive molecules including cholesterol. Some ABC transporters contribute to lowering of cerebral Aβ. Mechanisms suggested for ABC transporter-mediated lowering of brain Aβ, in addition to exporting of Aβ across the blood brain and blood cerebrospinal fluid barriers, include apolipoprotein E lipidation, microglial activation, decreased amyloidogenic processing of amyloid precursor protein, and restricting the entrance of Aβ into the brain. The ABC transporter superfamily in humans includes 49 proteins, eight of which have been suggested to reduce cerebral Aβ levels. This review discusses experimental approaches for increasing the expression of these ABC transporters, clinical applications of these approaches, changes in the expression and/or activity of these transporters in AD and transgenic mouse models of AD, and findings in the few clinical trials which have examined the effects of these approaches in patients with AD or mild cognitive impairment. The possibility that therapeutic upregulation of ABC transporters which promote clearance of cerebral Aβ may slow the clinical progression of AD merits further consideration.
Collapse
Affiliation(s)
- David A. Loeffler
- Department of Neurology, Beaumont Research Institute, Corewell Health, Royal Oak, MI, United States
| |
Collapse
|
4
|
Litvinchuk A, Suh JH, Guo JL, Lin K, Davis SS, Bien-Ly N, Tycksen E, Tabor GT, Remolina Serrano J, Manis M, Bao X, Lee C, Bosch M, Perez EJ, Yuede CM, Cashikar AG, Ulrich JD, Di Paolo G, Holtzman DM. Amelioration of Tau and ApoE4-linked glial lipid accumulation and neurodegeneration with an LXR agonist. Neuron 2024; 112:384-403.e8. [PMID: 37995685 PMCID: PMC10922706 DOI: 10.1016/j.neuron.2023.10.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 09/13/2023] [Accepted: 10/18/2023] [Indexed: 11/25/2023]
Abstract
Apolipoprotein E (APOE) is a strong genetic risk factor for late-onset Alzheimer's disease (LOAD). APOE4 increases and APOE2 decreases risk relative to APOE3. In the P301S mouse model of tauopathy, ApoE4 increases tau pathology and neurodegeneration when compared with ApoE3 or the absence of ApoE. However, the role of ApoE isoforms and lipid metabolism in contributing to tau-mediated degeneration is unknown. We demonstrate that in P301S tau mice, ApoE4 strongly promotes glial lipid accumulation and perturbations in cholesterol metabolism and lysosomal function. Increasing lipid efflux in glia via an LXR agonist or Abca1 overexpression strongly attenuates tau pathology and neurodegeneration in P301S/ApoE4 mice. We also demonstrate reductions in reactive astrocytes and microglia, as well as changes in cholesterol biosynthesis and metabolism in glia of tauopathy mice in response to LXR activation. These data suggest that promoting efflux of glial lipids may serve as a therapeutic approach to ameliorate tau and ApoE4-linked neurodegeneration.
Collapse
Affiliation(s)
- Alexandra Litvinchuk
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Jung H Suh
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA
| | - Jing L Guo
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA
| | - Karin Lin
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA
| | - Sonnet S Davis
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA
| | - Nga Bien-Ly
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA
| | - Eric Tycksen
- Genome Technology Access Center, McDonnell Genome Institute, St. Louis, MO 63110, USA
| | - G Travis Tabor
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Javier Remolina Serrano
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Melissa Manis
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Xin Bao
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Choonghee Lee
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Megan Bosch
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Enmanuel J Perez
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Carla M Yuede
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Anil G Cashikar
- Department of Psychiatry, Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Jason D Ulrich
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63108, USA
| | | | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63108, USA.
| |
Collapse
|
5
|
Zheng M, Ye H, Yang X, Shen L, Dang X, Liu X, Gong Y, Wu Q, Wang L, Ge X, Fang X, Hou B, Zhang P, Tang R, Zheng K, Huang XF, Yu Y. Probiotic Clostridium butyricum ameliorates cognitive impairment in obesity via the microbiota-gut-brain axis. Brain Behav Immun 2024; 115:565-587. [PMID: 37981012 DOI: 10.1016/j.bbi.2023.11.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 11/06/2023] [Accepted: 11/13/2023] [Indexed: 11/21/2023] Open
Abstract
Obesity is a risk factor for cognitive dysfunction and neurodegenerative disease, including Alzheimer's disease (AD). The gut microbiota-brain axis is altered in obesity and linked to cognitive impairment and neurodegenerative disorders. Here, we targeted obesity-induced cognitive impairment by testing the impact of the probiotic Clostridium butyricum, which has previously shown beneficial effects on gut homeostasis and brain function. Firstly, we characterized and analyzed the gut microbial profiles of participants with obesity and the correlation between gut microbiota and cognitive scores. Then, using an obese mouse model induced by a Western-style diet (high-fat and fiber-deficient diet), the effects of Clostridium butyricum on the microbiota-gut-brain axis and hippocampal cognitive function were evaluated. Finally, fecal microbiota transplantation was performed to assess the functional link between Clostridium butyricum remodeling gut microbiota and hippocampal synaptic protein and cognitive behaviors. Our results showed that participants with obesity had gut microbiota dysbiosis characterized by an increase in phylum Proteobacteria and a decrease in Clostridium butyricum, which were closely associated with cognitive decline. In diet-induced obese mice, oral Clostridium butyricum supplementation significantly alleviated cognitive impairment, attenuated the deficit of hippocampal neurite outgrowth and synaptic ultrastructure, improved hippocampal transcriptome related to synapses and dendrites; a comparison of the effects of Clostridium butyricum in mice against human AD datasets revealed that many of the genes changes in AD were reversed by Clostridium butyricum; concurrently, Clostridium butyricum also prevented gut microbiota dysbiosis, colonic barrier impairment and inflammation, and attenuated endotoxemia. Importantly, fecal microbiota transplantation from donor-obese mice with Clostridium butyricum supplementation facilitated cognitive variables and colonic integrity compared with from donor obese mice, highlighting that Clostridium butyricum's impact on cognitive function is largely due to its ability to remodel gut microbiota. Our findings provide the first insights into the neuroprotective effects of Clostridium butyricum on obesity-associated cognitive impairments and neurodegeneration via the gut microbiota-gut-brain axis.
Collapse
Affiliation(s)
- Mingxuan Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Huaiyu Ye
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Xiaoying Yang
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Lijun Shen
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Xuemei Dang
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Xiaoli Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Yuying Gong
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Qingyuan Wu
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Li Wang
- Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang 110033, China
| | - Xing Ge
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Xiaoli Fang
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Jiangsu 221004, China
| | - Benchi Hou
- Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang 110033, China
| | - Peng Zhang
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Renxian Tang
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Kuiyang Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; National Experimental Demonstration Center for Basic Medicine Education, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Xu-Feng Huang
- Illawarra Health and Medical Research Institute (IHMRI) and School of Medicine, University of Wollongong, NSW 2522, Australia
| | - Yinghua Yu
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China.
| |
Collapse
|
6
|
Alnaaim SA, Al-Kuraishy HM, Alexiou A, Papadakis M, Saad HM, Batiha GES. Role of Brain Liver X Receptor in Parkinson's Disease: Hidden Treasure and Emerging Opportunities. Mol Neurobiol 2024; 61:341-357. [PMID: 37606719 PMCID: PMC10791998 DOI: 10.1007/s12035-023-03561-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 08/01/2023] [Indexed: 08/23/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease due to the degeneration of dopaminergic neurons (DNs) in the substantia nigra (SN). The liver X receptor (LXR) is involved in different neurodegenerative diseases. Therefore, the objective of the present review was to clarify the possible role of LXR in PD neuropathology. LXRs are the most common nuclear receptors of transcription factors that regulate cholesterol metabolism and have pleiotropic effects, including anti-inflammatory effects and reducing intracellular cholesterol accumulation. LXRs are highly expressed in the adult brain and act as endogenous sensors for intracellular cholesterol. LXRs have neuroprotective effects against the development of neuroinflammation in different neurodegenerative diseases by inhibiting the expression of pro-inflammatory cytokines. LXRs play an essential role in mitigating PD neuropathology by reducing the expression of inflammatory signaling pathways, neuroinflammation, oxidative stress, mitochondrial dysfunction, and enhancement of BDNF signaling.In conclusion, LXRs, through regulating brain cholesterol homeostasis, may be effectual in PD. Also, inhibition of node-like receptor pyrin 3 (NLRP3) inflammasome and nuclear factor kappa B (NF-κB) by LXRs could effectively prevent neuroinflammation in PD. Taken together, LXRs play a crucial role in PD neuropathology by inhibiting neuroinflammation and associated degeneration of DNs.
Collapse
Affiliation(s)
- Saud A Alnaaim
- Clinical Neurosciences Department, College of Medicine, King Faisal University, Hofuf, Saudi Arabia
| | - Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Therapeutic Medicine, College of Medicine, ALmustansiriyiah University, Baghdad, 14132, Iraq
| | - Athanasios Alexiou
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW, 2770, Australia
- AFNP Med, 1030, Wien, Austria
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, Heusnerstrasse 40, University of Witten-Herdecke, 42283, Wuppertal, Germany.
| | - Hebatallah M Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Marsa Matruh, 51744, Egypt
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, AlBeheira, 22511, Egypt
| |
Collapse
|
7
|
Vacondio D, Nogueira Pinto H, Coenen L, Mulder IA, Fontijn R, van Het Hof B, Fung WK, Jongejan A, Kooij G, Zelcer N, Rozemuller AJ, de Vries HE, de Wit NM. Liver X receptor alpha ensures blood-brain barrier function by suppressing SNAI2. Cell Death Dis 2023; 14:781. [PMID: 38016947 PMCID: PMC10684660 DOI: 10.1038/s41419-023-06316-8] [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: 07/04/2023] [Revised: 11/09/2023] [Accepted: 11/15/2023] [Indexed: 11/30/2023]
Abstract
In Alzheimer's disease (AD) more than 50% of the patients are affected by capillary cerebral amyloid-angiopathy (capCAA), which is characterized by localized hypoxia, neuro-inflammation and loss of blood-brain barrier (BBB) function. Moreover, AD patients with or without capCAA display increased vessel number, indicating a reactivation of the angiogenic program. The molecular mechanism(s) responsible for BBB dysfunction and angiogenesis in capCAA is still unclear, preventing a full understanding of disease pathophysiology. The Liver X receptor (LXR) family, consisting of LXRα and LXRβ, was reported to inhibit angiogenesis and particularly LXRα was shown to secure BBB stability, suggesting a major role in vascular function. In this study, we unravel the regulatory mechanism exerted by LXRα to preserve BBB integrity in human brain endothelial cells (BECs) and investigate its role during pathological conditions. We report that LXRα ensures BECs identity via constitutive inhibition of the transcription factor SNAI2. Accordingly, deletion of brain endothelial LXRα is associated with impaired DLL4-NOTCH signalling, a critical signalling pathway involved in vessel sprouting. A similar response was observed when BECs were exposed to hypoxia, with concomitant LXRα decrease and SNAI2 increase. In support of our cell-based observations, we report a general increase in vascular SNAI2 in the occipital cortex of AD patients with and without capCAA. Importantly, SNAI2 strongly associated with vascular amyloid-beta deposition and angiopoietin-like 4, a marker for hypoxia. In hypoxic capCAA vessels, the expression of LXRα may decrease leading to an increased expression of SNAI2, and consequently BECs de-differentiation and sprouting. Our findings indicate that LXRα is essential for BECs identity, thereby securing BBB stability and preventing aberrant angiogenesis. These results uncover a novel molecular pathway essential for BBB identity and vascular homeostasis providing new insights on the vascular pathology affecting AD patients.
Collapse
Affiliation(s)
- D Vacondio
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, De Boelelaan 1108, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - H Nogueira Pinto
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, De Boelelaan 1108, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - L Coenen
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, De Boelelaan 1108, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Amsterdam, the Netherlands
- Biomedical Primate Research Centre, Department of Neurobiology and Aging, Rijswijk, the Netherlands
| | - I A Mulder
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, De Boelelaan 1108, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Amsterdam, the Netherlands
- Amsterdam UMC location University of Amsterdam, Department of Biomedical Engineering and Physics, Meibergdreef 9, Amsterdam, the Netherlands
| | - R Fontijn
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, De Boelelaan 1108, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - B van Het Hof
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, De Boelelaan 1108, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - W K Fung
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, De Boelelaan 1108, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - A Jongejan
- Amsterdam UMC location University of Amsterdam, Epidemiology and Data Science, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam Public Health, Methodology, Amsterdam, The Netherlands
- Amsterdam Infection and Immunity, Inflammatory Diseases, Amsterdam, The Netherlands
| | - G Kooij
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, De Boelelaan 1108, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - N Zelcer
- Amsterdam UMC location University of Amsterdam Department of Medical Biochemistry, Meibergdreef 9, Amsterdam, the Netherlands
- Amsterdam UMC location University of Amsterdam, Cardiovascular Sciences and Gastroenterology and Metabolism, Meibergdreef 9, Amsterdam, the Netherlands
| | - A J Rozemuller
- Amsterdam Neuroscience, Amsterdam, the Netherlands
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Pathology, De Boelelaan 1117, Amsterdam, the Netherlands
| | - H E de Vries
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, De Boelelaan 1108, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - N M de Wit
- Amsterdam UMC location Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, De Boelelaan 1108, Amsterdam, the Netherlands.
- Amsterdam Neuroscience, Amsterdam, the Netherlands.
| |
Collapse
|
8
|
Babalola JA, Lang M, George M, Stracke A, Tam-Amersdorfer C, Itxaso I, Lucija D, Tadic J, Schilcher I, Loeffler T, Flunkert S, Prokesch M, Leitinger G, Lass A, Hutter-Paier B, Panzenboeck U, Hoefler G. Astaxanthin enhances autophagy, amyloid beta clearance and exerts anti-inflammatory effects in in vitro models of Alzheimer's disease-related blood brain barrier dysfunction and inflammation. Brain Res 2023; 1819:148518. [PMID: 37579986 DOI: 10.1016/j.brainres.2023.148518] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/22/2023] [Accepted: 08/02/2023] [Indexed: 08/16/2023]
Abstract
Defective degradation and clearance of amyloid-β as well as inflammation per se are crucial players in the pathology of Alzheimer's disease (AD). A defective transport across the blood-brain barrier is causative for amyloid-β (Aβ) accumulation in the brain, provoking amyloid plaque formation. Using primary porcine brain capillary endothelial cells and murine organotypic hippocampal slice cultures as in vitro models of AD, we investigated the effects of the antioxidant astaxanthin (ASX) on Aβ clearance and neuroinflammation. We report that ASX enhanced the clearance of misfolded proteins in primary porcine brain capillary endothelial cells by inducing autophagy and altered the Aβ processing pathway. We observed a reduction in the expression levels of intracellular and secreted amyloid precursor protein/Aβ accompanied by an increase in ABC transporters ABCA1, ABCG1 as well as low density lipoprotein receptor-related protein 1 mRNA levels. Furthermore, ASX treatment increased autophagic flux as evidenced by increased lipidation of LC3B-II as well as reduced protein expression of phosphorylated S6 ribosomal protein and mTOR. In LPS-stimulated brain slices, ASX exerted anti-inflammatory effects by reducing the secretion of inflammatory cytokines while shifting microglia polarization from M1 to M2 phenotype. Our data suggest ASX as potential therapeutic compound ameliorating AD-related blood brain barrier impairment and inflammation.
Collapse
Affiliation(s)
| | - Magdalena Lang
- Otto Loewi Research Center, Division of Immunology, Medical University of Graz, Austria
| | - Meekha George
- Department of Obstetrics and Gynaecology, Medical University of Graz, Austria
| | - Anika Stracke
- Otto Loewi Research Center, Division of Immunology, Medical University of Graz, Austria
| | | | | | | | - Jelena Tadic
- Institute of Molecular Biosciences, University of Graz, Austria
| | | | | | | | | | - Gerd Leitinger
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Austria
| | - Achim Lass
- Institute of Molecular Biosciences, University of Graz, Austria
| | | | - Ute Panzenboeck
- Otto Loewi Research Center, Division of Immunology, Medical University of Graz, Austria
| | - Gerald Hoefler
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Austria.
| |
Collapse
|
9
|
Paseban T, Alavi MS, Etemad L, Roohbakhsh A. The role of the ATP-Binding Cassette A1 (ABCA1) in neurological disorders: a mechanistic review. Expert Opin Ther Targets 2023; 27:531-552. [PMID: 37428709 DOI: 10.1080/14728222.2023.2235718] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 07/09/2023] [Indexed: 07/12/2023]
Abstract
INTRODUCTION Cholesterol homeostasis is critical for normal brain function. It is tightly controlled by various biological elements. ATP-binding cassette transporter A1 (ABCA1) is a membrane transporter that effluxes cholesterol from cells, particularly astrocytes, into the extracellular space. The recent studies pertaining to ABCA1's role in CNS disorders were included in this study. AREAS COVERED In this comprehensive literature review, preclinical and human studies showed that ABCA1 has a significant role in the following diseases or disorders: Alzheimer's disease, Parkinson's disease, Huntington's disease, multiple sclerosis, neuropathy, anxiety, depression, psychosis, epilepsy, stroke, and brain ischemia and trauma. EXPERT OPINION ABCA1 via modulating normal and aberrant brain functions such as apoptosis, phagocytosis, BBB leakage, neuroinflammation, amyloid β efflux, myelination, synaptogenesis, neurite outgrowth, and neurotransmission promotes beneficial effects in aforementioned diseases. ABCA1 is a key molecule in the CNS. By boosting its expression or function, some CNS disorders may be resolved. In preclinical studies, liver X receptor agonists have shown promise in treating CNS disorders via ABCA1 and apoE enhancement.
Collapse
Affiliation(s)
- Tahere Paseban
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohaddeseh Sadat Alavi
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Leila Etemad
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Roohbakhsh
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
10
|
Na Y, Ke L, Jie Z, Jinping W, Tao M, Jie Z, Liu Y, Yueqin Z. Amelioration of Cholesterol Rich diet-induced Impaired Cognition in AD Transgenic Mice by an LXR Agonist TO901317 Is Associated with the Activation of the LXR-β-RXR-α-ABCA1 Transmembrane Transport System and Improving the Composition of Lipid Raft. Exp Aging Res 2023; 49:214-225. [PMID: 35792710 DOI: 10.1080/0361073x.2022.2095605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 06/26/2022] [Indexed: 11/04/2022]
Abstract
BACKGROUND It has been reported that LXR agonist can inhibit Aβ generation and alleviate Aβ-induced various adverse reactions in vivo and in vitro experiments, but the mechanisms have not been clarified. The study aimed to observe the effect of LXR agonist TO901317 on the cognitive function of AD transgenic mice fed with cholesterol-rich diet (CRD), and to explore the possible mechanism. Methods: 32 male 6-month-old double transgenic AD mice were enrolled and randomly divided into 4 groups: control (normal diet) group, CRD treatment group, TO901317 treatment group and GSK2033 treatment group. After 3 month, Morris water maze was for the changes of spatial exploration and memory ability; ELISA was for detecting the production of Aβ42 in the brain; the concentration of total cholesterol (TC), low density lipoprotein (LDL) and high density lipoprotein (HDL) in serum were detected by cholesterol enzyme colorimetry; Finally, the expression of LXR-β, RXR-α, ABCA1, caveolin-1, BACE1 and APP at protein level in the brains was measured by Western blotting. RESULTS Compared with the control group, the learning, memory ability and spatial exploration ability of the mice were more significantly serious in the CRD group (P<0.05); The contents of TC and LDL in the serum and the production of Aβ42 in the brains were significantly increased (P<0.05), but HDL was remarkably decreased (P<0.05); The protein levels of LXR-β, RXR-α and ABCA1 were also significantly decreased (P<0.05); The expression of caveolin-1, APP and BACE1 were evidently increased (P<0.05). However, after treatment with TO901317, the impaired learning and memory and spatial exploration ability of the mice were significantly improved (P<0.05); The contents of TC and LDL in serum and the production of Aβ42 in the brains were significantly decreased (P<0.05), but HLD was increased (P<0.05); The protein levels of LXR-β, RXR-α, ABCA1were all significantly increased (P<0.05), while, the expression of caveolin-1, APP and BACE1 were all significantly decreased (P<0.05). All the changes were reversed by GSK2033 (P<0.05). CONCLUSIONS TO901317 attenuated the more serious impairment of spatial exploration, learning and memory in transgenic AD mice induced by CRD, and the mechanism may be that TO901317 could activate the LXR-β/RXR-α/ABCA1 transmembrane transport system, promote the cholesterol efflux, and decreased caveolin-1, APP and BACE1, further reduce Aβ42 in the brains.
Collapse
Affiliation(s)
- Yang Na
- Chongqing Emergence Medical Center, Chongqing University Central Hopital, Chongqing, Sichuan, China
| | - Lin Ke
- Chongqing Emergence Medical Center, Chongqing University Central Hopital, Chongqing, Sichuan, China
| | - Zhang Jie
- Chongqing Emergence Medical Center, Chongqing University Central Hopital, Chongqing, Sichuan, China
| | - Wang Jinping
- Chongqing Emergence Medical Center, Chongqing University Central Hopital, Chongqing, Sichuan, China
| | - Meng Tao
- Chongqing Emergence Medical Center, Chongqing University Central Hopital, Chongqing, Sichuan, China
| | - Zhu Jie
- Chongqing Emergence Medical Center, Chongqing University Central Hopital, Chongqing, Sichuan, China
| | - Yang Liu
- Chongqing Emergence Medical Center, Chongqing University Central Hopital, Chongqing, Sichuan, China
| | - Zhou Yueqin
- Chongqing Emergence Medical Center, Chongqing University Central Hopital, Chongqing, Sichuan, China
| |
Collapse
|
11
|
Luo Y, Tan X, Zhang X, Li Y, Huang J, Deng Y. Effect of liver X receptor agonist TO901317 on cognitive function in APP/PS1 double transgenic mice with Alzheimer 's disease and the underlying mechanism. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2022; 47:1324-1331. [PMID: 36411683 PMCID: PMC10930358 DOI: 10.11817/j.issn.1672-7347.2022.210770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Indexed: 06/16/2023]
Abstract
OBJECTIVES The liver X receptors (LXRs) are members of the nuclear hormone receptor superfamily, and LXR-β is an important receptor for cholesterol content in brain cells. LXR-β/retinoic X receptor (RXR-α)/ATP binding cassette transporter A1 (ABCA1) cholesterol transmembrane transport system is closely related to the occurrence and development of Alzheimer's disease (AD). LXR agonist TO901317 can affect the accumulation of β- amyloid protein in the brain tissue of APP/PS1 double transgenic AD mice. However, the molecular mechanism is not clarified in detail. This study aims to evaluate the effects of LXR agonist TO901317 on the cognitive function of AD mice fed with high cholesterol diet, and to explore its possible mechanism from the perspective of cholesterol metabolism. METHODS Twenty four male 6-month-old APP/PS1 double transgenic AD mice were randomly divided into 4 groups, 6 mice in each group: a control group (fed with normal diet), a cholesterol rich diet (CRD) group, a TO901317 group (fed with CRD combined with TO901317), and a GSK2033 group (fed with CRD combined with TO901317 and LXR antagonist GSK2033). The mice were fed with pellet feed made of high cholesterol feed, mixed with lard, egg yolk powder, and cod liver oil twice a day. TO901317 and GSK2033 were dissolved and diluted to a final concentration at 0.03%. The drugs were given to the mice daily through gastric tube according to their body weight. Meanwhile, the mice in the drug group were fed with high cholesterol diet . After feeding for 3 months, Morris water maze was used to observe the changes of spatial exploration and memory ability of AD mice in each group. The contents of TC, LDL, and HDL in serum of mice in each group were detected by cholesterol enzyme colorimetry, and the differences among the groups were compared. The expression of Aβ42 in the brain of AD mice was detected by ELISA. Western blotting was used to detect the protein levels of LXR-β, RXR-α, ABCA1, and Caveolin-1 in the brain of each group. RESULTS Morris water maze results showed that the times, distance and the duration of mice crossing the platform in the CRD group were significantly decreased compared with the control group (all P<0.05), while these three figures in TO901317 group were significantly increased compared with the CRD group (all P<0.05). Compared with the TO901317 group, there was a decrease of these figures in the GSK2033 group (all P<0.05). The serum TC and LDL levels in the CRD group were significantly higher than those in the control group, while HDL levels were significantly lower (all P<0.001). The figures of the TC and LDL contents level in the TO901317 group were lower than those in the CRD group, while HDL levels were higher (all P<0.001). Compared with TO901317 group, the contents of the TC and LDL in GSK2033 group were significantly increased, while HDL content was significantly decreased (all P<0.001). ELISA results showed that the production of Aβ42 peptides in the brain of CRD group was the highest while the content in the TO901317 group was significantly decreased (P<0.001), which was the lowest among the groups. The figure in the control group was close to the GSK2033 group. Western blotting results showed that the protein levels of LXR-β, RXR-α, and ABCA1 in the CRD group were significantly decreased compared with the control group, but the protein level of Caveolin-1 was increased (all P<0.01). After TO901317 treatment, the protein levels of LXR-β, RXR-α and ABCA1 were significantly increased, while the protein level of Caveolin-1 was decreased partially (all P<0.001). In the GSK2033 group, the effect of TO901317 on AD mice was partially reversed by GSK2033. Compared to TO901317 group, the protein levels of LXR-β, RXR-α, and ABCA1 showed a decrease trend, while the protein level of Caveolin-1 showed an increase state (all P<0.05). CONCLUSIONS High cholesterol diet leads to severer spatial exploration, learning and memory impairment in transgenic AD mice, while the LXR agonist TO901317 attenuates this effect. The mechanism may be that TO901317 promotes cholesterol efflux by activating LXR-β/RXR-α/ABCA1 transmembrane transport system, reduces the expression of Caveolin-1, improves the composition of lipid raft, and ultimately reduces the production of Aβ42 in the brain.
Collapse
Affiliation(s)
- Yingmao Luo
- Department of Gerontology, Chongqing Mental Health Center, Chongqing 400010.
| | - Xiaolin Tan
- Department of Gerontology, Chongqing Mental Health Center, Chongqing 400010
| | - Xiong Zhang
- Neuroscience Research Center, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yuan Li
- Department of Gerontology, Chongqing Mental Health Center, Chongqing 400010
| | - Jie Huang
- Department of Gerontology, Chongqing Mental Health Center, Chongqing 400010
| | - Yu Deng
- Department of Gerontology, Chongqing Mental Health Center, Chongqing 400010.
| |
Collapse
|
12
|
Pedrini S, Doecke JD, Hone E, Wang P, Thota R, Bush AI, Rowe CC, Dore V, Villemagne VL, Ames D, Rainey‐Smith S, Verdile G, Sohrabi HR, Raida MR, Taddei K, Gandy S, Masters CL, Chatterjee P, Martins R. Plasma high-density lipoprotein cargo is altered in Alzheimer's disease and is associated with regional brain volume. J Neurochem 2022; 163:53-67. [PMID: 36000528 PMCID: PMC9804612 DOI: 10.1111/jnc.15681] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 07/12/2022] [Accepted: 07/22/2022] [Indexed: 01/05/2023]
Abstract
Cholesterol levels have been repeatedly linked to Alzheimer's Disease (AD), suggesting that high levels could be detrimental, but this effect is likely attributed to Low-Density Lipoprotein (LDL) cholesterol. On the other hand, High-Density Lipoproteins (HDL) cholesterol levels have been associated with reduced brain amyloidosis and improved cognitive function. However, recent findings have suggested that HDL-functionality, which depends upon the HDL-cargo proteins associated with HDL, rather than HDL levels, appears to be the key factor, suggesting a quality over quantity status. In this report, we have assessed the HDL-cargo (Cholesterol, ApoA-I, ApoA-II, ApoC-I, ApoC-III, ApoD, ApoE, ApoH, ApoJ, CRP, and SAA) in stable healthy control (HC), healthy controls who will convert to MCI/AD (HC-Conv) and AD patients (AD). Compared to HC we observed an increased cholesterol/ApoA-I ratio in AD and HC-Conv, as well as an increased ApoD/ApoA-I ratio and a decreased ApoA-II/ApoA-I ratio in AD. Higher cholesterol/ApoA-I ratio was also associated with lower cortical grey matter volume and higher ventricular volume, while higher ApoA-II/ApoA-I and ApoJ/ApoA-I ratios were associated with greater cortical grey matter volume (and for ApoA-II also with greater hippocampal volume) and smaller ventricular volume. Additionally, in a clinical status-independent manner, the ApoE/ApoA-I ratio was significantly lower in APOE ε4 carriers and lowest in APOE ε4 homozygous. Together, these data indicate that in AD patients the composition of HDL is altered, which may affect HDL functionality, and such changes are associated with altered regional brain volumetric data.
Collapse
Affiliation(s)
- Steve Pedrini
- School of Medical SciencesEdith Cowan UniversityJoondalupWestern AustraliaAustralia,CRC for Mental HealthMelbourneVictoriaAustralia
| | - James D. Doecke
- Australian E‐Health Research CentreCSIROBrisbaneQueenslandAustralia
| | - Eugene Hone
- School of Medical SciencesEdith Cowan UniversityJoondalupWestern AustraliaAustralia,CRC for Mental HealthMelbourneVictoriaAustralia
| | - Penghao Wang
- College of Science, Health, Engineering and EducationMurdoch UniversityMurdochWestern AustraliaAustralia
| | - Rohith Thota
- Faculty of Medicine, Health and Human Sciences, Department of Biomedical SciencesMacquarie UniversitySydneyNew South WalesAustralia
| | - Ashley I. Bush
- CRC for Mental HealthMelbourneVictoriaAustralia,The Florey Institute, The University of MelbourneParkvilleVictoriaAustralia
| | - Christopher C. Rowe
- Department of Nuclear Medicine and Centre for PETAustin HealthHeidelbergVictoriaAustralia
| | - Vincent Dore
- Department of Nuclear Medicine and Centre for PETAustin HealthHeidelbergVictoriaAustralia
| | | | - David Ames
- National Ageing Research InstituteParkvilleVictoriaAustralia,University of Melbourne Academic unit for Psychiatry of Old AgeSt George's HospitalKewVictoriaAustralia
| | - Stephanie Rainey‐Smith
- School of Medical SciencesEdith Cowan UniversityJoondalupWestern AustraliaAustralia,Centre for Healthy Ageing, Health Futures InstituteMurdoch UniversityMurdochWestern AustraliaAustralia
| | - Giuseppe Verdile
- Curtin Medical SchoolCurtin UniversityBentleyWestern AustraliaAustralia,Curtin Health Innovation Research InstituteCurtin UniversityBentleyWestern AustraliaAustralia
| | - Hamid R. Sohrabi
- Centre for Healthy Ageing, Health Futures InstituteMurdoch UniversityMurdochWestern AustraliaAustralia
| | - Manfred R. Raida
- Life Science Institute, Singapore Lipidomics IncubatorNational University of SingaporeSingapore CitySingapore
| | - Kevin Taddei
- School of Medical SciencesEdith Cowan UniversityJoondalupWestern AustraliaAustralia,CRC for Mental HealthMelbourneVictoriaAustralia
| | - Sam Gandy
- Department of NeurologyIcahn School of Medicine at Mount SinaiNew York CityNew YorkUSA
| | - Colin L. Masters
- The Florey Institute, The University of MelbourneParkvilleVictoriaAustralia
| | - Pratishtha Chatterjee
- Faculty of Medicine, Health and Human Sciences, Department of Biomedical SciencesMacquarie UniversitySydneyNew South WalesAustralia
| | - Ralph N. Martins
- School of Medical SciencesEdith Cowan UniversityJoondalupWestern AustraliaAustralia,CRC for Mental HealthMelbourneVictoriaAustralia,Faculty of Medicine, Health and Human Sciences, Department of Biomedical SciencesMacquarie UniversitySydneyNew South WalesAustralia,School of Psychiatry and Clinical NeurosciencesUniversity of Western AustraliaCrawleyWestern AustraliaAustralia
| | | |
Collapse
|
13
|
Liver X Receptor Regulation of Glial Cell Functions in the CNS. Biomedicines 2022; 10:biomedicines10092165. [PMID: 36140266 PMCID: PMC9496004 DOI: 10.3390/biomedicines10092165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/19/2022] [Accepted: 08/29/2022] [Indexed: 11/22/2022] Open
Abstract
In this review, we discuss the role of liver X receptors (LXRs) in glial cells (microglia, oligodendrocytes and astrocytes) in the central nervous system (CNS). LXRs are oxysterol-activated nuclear receptors that, in adults, regulate genes involved in cholesterol homeostasis, the modulation of inflammatory responses and glutamate homeostasis. The study of LXR knockout mice has revealed that LXRβ plays a key role in maintaining the health of dopaminergic neurons in the substantia nigra, large motor neurons in the spinal cord and retinal ganglion cells in the eye. In the peripheral nervous system (PNS), LXRβ is responsible for the health of the spiral ganglion neurons (SGNs) in the cochlea. In addition, LXRs are essential for the homeostasis of the cerebrospinal fluid (CSF), and in LXRαβ−/− mice, the lateral ventricles are empty and lined with lipid-laden cells. As LXRαβ−/− mice age, lipid vacuoles accumulate in astrocytes surrounding blood vessels. By seven months of age, motor coordination becomes impaired, and there is a loss of motor neurons in the spinal cord of LXRβ−/− mice. During development, migration of neurons in the cortex and cerebellum is retarded in LXRβ−/− mice. Since LXRs are not expressed in dopaminergic or motor neurons in adult mice, the neuroprotective effects of LXRs appear to come from LXRs in glial cells where they are expressed. However, despite the numerous neurological deficits in LXR−/− rodents, multiple sclerosis has the clear distinction of being the only human neurodegenerative disease in which defective LXR signaling has been identified. In this review, we summarize the regulation and functions of LXRs in glial cells and analyze how targeting LXRs in glial cells might, in the future, be used to treat neurodegenerative diseases and, perhaps, disorders caused by aberrant neuronal migration during development.
Collapse
|
14
|
Huo C, Chen MH, Hour TC, Huang LC, Fong YO, Kuo YY, Yang YH, Chuu CP. Application of Micro-Western Array for Identifying Different Serum Protein Expression Profile among Healthy Control, Alzheimer’s Disease Patients and Patients’ Adult Children. Brain Sci 2022; 12:brainsci12091134. [PMID: 36138870 PMCID: PMC9496696 DOI: 10.3390/brainsci12091134] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 08/17/2022] [Accepted: 08/23/2022] [Indexed: 12/03/2022] Open
Abstract
(1) Background: Alzheimer’s disease (AD) is the most common form of dementia. Increased levels of inflammatory proteins have been observed in brain and plasma samples of AD patients; however, it is not clear if other serum proteins correlate to the development or disease progression of AD. (2) Methods: Micro-Western Array (MWA) is a high-throughput antibody-based proteomics system which allows detection of the expression levels of 24–96 different proteins within 6–30 samples simultaneously. We applied MWA to explore potential serum protein biomarkers correlated to the development and progression of AD by examining the difference in serum protein profile of 31 healthy control (HC), 30 patients with AD and 30 patients’ adult children (ACS). (3) Results: Compared to HC, AD and ACS express similar pattern of serum proteins, including higher protein levels of ABCA1, ABCG1, SREBP1 and LXRβ but lower protein levels of ApoD, ApoE, ApoH, c_Myc, COX2 and Hippo-YAP signaling proteins. AD patients had higher serum levels of ABCG1, ApoD, ApoH, COX2, LXRα and YAP, but lower levels of ABCA1, ApoE, c_Myc, LATS1, MST1, MST2, Nanog, NFκB_p50, PPARγ and SREBP2, as compared to ACS. Pearson’s correlation analysis revealed that the protein expression level of ApoE, c_Myc, LATS1, MST2, NFκB p50, PPARγ and SREBP1 was negatively correlated to age, while that of ApoE, c_Myc, LATS1, MST1, MST2, Nanog, NFκB p50 and PPARγ was positively correlated to age. (4) Conclusions: We identified a group of serum proteins which may correlate to disease progression of AD and can be potential diagnostic serum protein biomarkers.
Collapse
Affiliation(s)
- Chieh Huo
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Ming-Hui Chen
- Neuroscience Research Center, Kaohsiung Medical University, Kaohsiung City 80708, Taiwan
| | - Tzyh-Chyuan Hour
- Neuroscience Research Center, Kaohsiung Medical University, Kaohsiung City 80708, Taiwan
- Department of Biochemistry, School of Medicine, Kaohsiung Medical University, Kaohsiung City 80708, Taiwan
| | - Ling-Chun Huang
- Department of Biochemistry, School of Medicine, Kaohsiung Medical University, Kaohsiung City 80708, Taiwan
- Department of Neurology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung City 80145, Taiwan
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung City 80756, Taiwan
| | - Yi-On Fong
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung City 80756, Taiwan
| | - Ying-Yu Kuo
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Yuan-Han Yang
- Neuroscience Research Center, Kaohsiung Medical University, Kaohsiung City 80708, Taiwan
- Department of Neurology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung City 80145, Taiwan
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung City 80756, Taiwan
- School of Post-Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung City 80708, Taiwan
- Correspondence: (Y.-H.Y.); (C.-P.C.); Tel.: +886-7-3162-158 (Y.-H.Y.); +886-37-206-166 (ext. 37300) (C.-P.C.)
| | - Chih-Pin Chuu
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli 35053, Taiwan
- Department of Life Sciences, National Central University, Taoyuan City 32031, Taiwan
- PhD Program for Aging, Graduate Institute of Basic Medical Science, China Medical University, Taichung City 40402, Taiwan
- Biotechnology Center, National Chung Hsing University, Taichung City 40227, Taiwan
- Correspondence: (Y.-H.Y.); (C.-P.C.); Tel.: +886-7-3162-158 (Y.-H.Y.); +886-37-206-166 (ext. 37300) (C.-P.C.)
| |
Collapse
|
15
|
Rudajev V, Novotny J. Cholesterol as a key player in amyloid β-mediated toxicity in Alzheimer’s disease. Front Mol Neurosci 2022; 15:937056. [PMID: 36090253 PMCID: PMC9453481 DOI: 10.3389/fnmol.2022.937056] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 07/27/2022] [Indexed: 11/13/2022] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder that is one of the most devastating and widespread diseases worldwide, mainly affecting the aging population. One of the key factors contributing to AD-related neurotoxicity is the production and aggregation of amyloid β (Aβ). Many studies have shown the ability of Aβ to bind to the cell membrane and disrupt its structure, leading to cell death. Because amyloid damage affects different parts of the brain differently, it seems likely that not only Aβ but also the nature of the membrane interface with which the amyloid interacts, helps determine the final neurotoxic effect. Because cholesterol is the dominant component of the plasma membrane, it plays an important role in Aβ-induced toxicity. Elevated cholesterol levels and their regulation by statins have been shown to be important factors influencing the progression of neurodegeneration. However, data from many studies have shown that cholesterol has both neuroprotective and aggravating effects in relation to the development of AD. In this review, we attempt to summarize recent findings on the role of cholesterol in Aβ toxicity mediated by membrane binding in the pathogenesis of AD and to consider it in the broader context of the lipid composition of cell membranes.
Collapse
|
16
|
Bossaerts L, Cacace R, Van Broeckhoven C. The role of ATP-binding cassette subfamily A in the etiology of Alzheimer's disease. Mol Neurodegener 2022; 17:31. [PMID: 35477481 PMCID: PMC9044696 DOI: 10.1186/s13024-022-00536-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 04/01/2022] [Indexed: 11/12/2022] Open
Abstract
Background Alzheimer’s disease (AD) is the leading cause of dementia, clinically characterized by memory deficits and progressive cognitive decline. Despite decades of research effective therapies are lacking, and a large part of the genetic heritability remains unidentified. ABCA7 and ABCA1, members of the ATP-binding cassette subfamily A (ABCA), were identified as AD risk genes in genome-wide association studies. Nevertheless, genetic and/or functional studies propose a link between AD and two other members of the ABCA subclass, i.e., ABCA2 and ABCA5. Main body Changes in expression or dysfunction of these transporters were found to increase amyloid β levels. This might be related to the common role of ABCA transporters in cellular cholesterol homeostasis, for which a prominent role in AD development has been suggested. In this review, we provide a comprehensive overview and discussion on the contribution of the ABCA subfamily to the etiopathogenesis of AD. Conclusions A better understanding of the function and identification of disease-associated genetic variants in ABCA transporters can contribute to the development of novel therapeutic strategies for AD.
Collapse
Affiliation(s)
- Liene Bossaerts
- Neurodegenerative Brain Diseases Group, VIB Center for Molecular Neurology, Antwerp, Belgium
| | - Rita Cacace
- Neurodegenerative Brain Diseases Group, VIB Center for Molecular Neurology, Antwerp, Belgium
| | - Christine Van Broeckhoven
- Neurodegenerative Brain Diseases Group, VIB Center for Molecular Neurology, Antwerp, Belgium. .,Department of Biomedical Sciences, University of Antwerp - CDE, Universiteitsplein 1, B-2610, Antwerp, Belgium.
| |
Collapse
|
17
|
Wu M, Zhai Y, Liang X, Chen W, Lin R, Ma L, Huang Y, Zhao D, Liang Y, Zhao W, Fang J, Fang S, Chen Y, Wang Q, Li W. Connecting the Dots Between Hypercholesterolemia and Alzheimer’s Disease: A Potential Mechanism Based on 27-Hydroxycholesterol. Front Neurosci 2022; 16:842814. [PMID: 35464321 PMCID: PMC9021879 DOI: 10.3389/fnins.2022.842814] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 03/01/2022] [Indexed: 12/13/2022] Open
Abstract
Alzheimer’s disease (AD), the most common cause of dementia, is a complex and multifactorial disease involving genetic and environmental factors, with hypercholesterolemia considered as one of the risk factors. Numerous epidemiological studies have reported a positive association between AD and serum cholesterol levels, and experimental studies also provide evidence that elevated cholesterol levels accelerate AD pathology. However, the underlying mechanism of hypercholesterolemia accelerating AD pathogenesis is not clear. Here, we review the metabolism of cholesterol in the brain and focus on the role of oxysterols, aiming to reveal the link between hypercholesterolemia and AD. 27-hydroxycholesterol (27-OHC) is the major peripheral oxysterol that flows into the brain, and it affects β-amyloid (Aβ) production and elimination as well as influencing other pathogenic mechanisms of AD. Although the potential link between hypercholesterolemia and AD is well established, cholesterol-lowering drugs show mixed results in improving cognitive function. Nevertheless, drugs that target cholesterol exocytosis and conversion show benefits in improving AD pathology. Herbs and natural compounds with cholesterol-lowering properties also have a potential role in ameliorating cognition. Collectively, hypercholesterolemia is a causative risk factor for AD, and 27-OHC is likely a potential mechanism for hypercholesterolemia to promote AD pathology. Drugs that regulate cholesterol metabolism are probably beneficial for AD, but more research is needed to unravel the mechanisms involved in 27-OHC, which may lead to new therapeutic strategies for AD.
Collapse
Affiliation(s)
- Mingan Wu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yingying Zhai
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaoyi Liang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Weichun Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ruiyi Lin
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Linlin Ma
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yi Huang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Di Zhao
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yong Liang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wei Zhao
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiansong Fang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shuhuan Fang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yunbo Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qi Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
- *Correspondence: Qi Wang,
| | - Weirong Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
- Weirong Li,
| |
Collapse
|
18
|
Plucińska K, Mody N, Dekeryte R, Shearer K, Mcilroy GD, Delibegovic M, Platt B. High-fat diet exacerbates cognitive and metabolic abnormalities in neuronal BACE1 knock-in mice - partial prevention by Fenretinide. Nutr Neurosci 2022; 25:719-736. [PMID: 32862802 DOI: 10.1080/1028415x.2020.1806190] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Objective: The β-site APP-cleaving enzyme 1 (BACE1) is a rate-limiting step in β-amyloid (Aβ) production in Alzheimer's disease (AD) brains, but recent evidence suggests that BACE1 is also involved in metabolic regulation. Here, we aimed to assess the effects of highfat diet (HFD) on metabolic and cognitive phenotypes in the diabetic BACE1 knock-in mice (PLB4) and WT controls; we additionally examined whether these phenotypes can be normalized with a synthetic retinoid (Fenretinide, Fen) targeting weight loss.Methods: Five-month old male WT and PLB4 mice were fed either (1) control chow diet, (2) 45%-saturated fat diet (HFD), (3) HFD with 0.04% Fen (HFD + Fen) or (4) control chow diet with 0.04% Fen (Fen) for 10 weeks. We assessed basic metabolic parameters, circadian rhythmicity, spatial habituation (Phenotyper) and working memory (Y-maze). Hypothalami, forebrain and liver tissues were assessed using Western blots, qPCR and ELISAs.Results: HFD feeding drastically worsened metabolism and induced early mortality (-40%) in otherwise viable PLB4 mice. This was ameliorated by Fen, despite no effects on glucose intolerance. In HFD-fed WT mice, Fen reduced weight gain, glucose intolerance and hepatic steatosis. The physiological changes induced in WT and PLB4 mice by HFD (+/-Fen) were accompanied by enhanced cerebral astrogliosis, elevated PTP1B, phopsho-eIF2α and altered hypothalamic transcription of Bace1, Pomc and Mc4r. Behaviourally, HFD feeding exacerbated spatial memory deficits in PLB4 mice, which was prevented by Fen and linked with increased full-length APP, normalized brain Aβ*56 oligomerization and astrogliosis.Conclusions: HFD induces early mortality and worsened cognition in the Alzheimer's-like BACE1 mice- partial prevention was achieved with Fenretinide, without improvements in glucose homeostasis.
Collapse
Affiliation(s)
- Kaja Plucińska
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
- The Novo Nordisk Foundation Center for Basic Metabolic Research (CBMR), Integrative Physiology and Environmental Influences, University of Copenhagen, Copenhagen, Denmark
| | - Nimesh Mody
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Ruta Dekeryte
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Kirsty Shearer
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - George D Mcilroy
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
- The Rowett Institute, University of Aberdeen, Aberdeen, UK
| | - Mirela Delibegovic
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - Bettina Platt
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
19
|
Lewandowski CT, Laham MS, Thatcher GR. Remembering your A, B, C's: Alzheimer's disease and ABCA1. Acta Pharm Sin B 2022; 12:995-1018. [PMID: 35530134 PMCID: PMC9072248 DOI: 10.1016/j.apsb.2022.01.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/27/2021] [Accepted: 01/07/2022] [Indexed: 12/24/2022] Open
Abstract
The function of ATP binding cassette protein A1 (ABCA1) is central to cholesterol mobilization. Reduced ABCA1 expression or activity is implicated in Alzheimer's disease (AD) and other disorders. Therapeutic approaches to boost ABCA1 activity have yet to be translated successfully to the clinic. The risk factors for AD development and progression, including comorbid disorders such as type 2 diabetes and cardiovascular disease, highlight the intersection of cholesterol transport and inflammation. Upregulation of ABCA1 can positively impact APOE lipidation, insulin sensitivity, peripheral vascular and blood–brain barrier integrity, and anti-inflammatory signaling. Various strategies towards ABCA1-boosting compounds have been described, with a bias toward nuclear hormone receptor (NHR) agonists. These agonists display beneficial preclinical effects; however, important side effects have limited development. In particular, ligands that bind liver X receptor (LXR), the primary NHR that controls ABCA1 expression, have shown positive effects in AD mouse models; however, lipogenesis and unwanted increases in triglyceride production are often observed. The longstanding approach, focusing on LXRβ vs. LXRα selectivity, is over-simplistic and has failed. Novel approaches such as phenotypic screening may lead to small molecule NHR modulators that elevate ABCA1 function without inducing lipogenesis and are clinically translatable.
Collapse
|
20
|
Katsuki H. Nuclear receptors of NR1 and NR4 subfamilies in the regulation of microglial functions and pathology. Pharmacol Res Perspect 2021; 9:e00766. [PMID: 34676987 PMCID: PMC8532137 DOI: 10.1002/prp2.766] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/01/2021] [Accepted: 03/15/2021] [Indexed: 12/15/2022] Open
Abstract
This review provides an overview of researches on the NR1 and NR4 nuclear receptors involved in the regulation of microglial functions. Nuclear receptors are attractive candidates for drug targets in the therapies of the central nervous system disorders, because the activation of these receptors is expected to regulate the functions and the phenotypes of microglia, by controlling the expression of specific gene subsets and also by regulating the cellular signaling mechanisms in a nongenomic manner. Several members of NR1 nuclear receptor subfamily have been examined for their ability to regulate microglial functions. For example, stimulation of vitamin D receptor inhibits the production of pro-inflammatory factors and increases the production of anti-inflammatory cytokines. Similar regulatory actions of nuclear receptor ligands on inflammation-related genes have also been reported for other NR1 members such as retinoic acid receptors, peroxisome proliferator-activated receptors (PPARs), and liver X receptors (LXRs). In addition, stimulation of PPARγ and LXRs may also result in increased phagocytic activities of microglia. Consistent with these actions, the agonists at nuclear receptors of NR1 subfamily are shown to produce therapeutic effects on animal models of various neurological disorders such as experimental allergic encephalomyelitis, Alzheimer's disease, Parkinson's disease, and ischemic/hemorrhagic stroke. On the other hand, increasing lines of evidence suggest that the stimulation of NR4 subfamily members of nuclear receptors such as Nur77 and Nurr1 also regulates microglial functions and alleviates neuropathological events in several disease models. Further advancement of these research fields may prove novel therapeutic opportunities.
Collapse
Affiliation(s)
- Hiroshi Katsuki
- Department of Chemico‐Pharmacological SciencesGraduate School of Pharmaceutical SciencesKumamoto UniversityKumamotoJapan
| |
Collapse
|
21
|
Chiang MC, Nicol CJB, Chen SJ, Huang RN. TO901317 activation of LXR-dependent pathways mitigate amyloid-beta peptide-induced neurotoxicity in 3D human neural stem cell culture scaffolds and AD mice. Brain Res Bull 2021; 178:57-68. [PMID: 34801648 DOI: 10.1016/j.brainresbull.2021.11.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 11/04/2021] [Accepted: 11/11/2021] [Indexed: 11/02/2022]
Abstract
Alzheimer's disease (AD) is the major cause of neurodegeneration worldwide and is characterized by the accumulation of amyloid beta (Aβ) in the brain, which is associated with neuronal loss and cognitive impairment. Liver X receptor (LXR), a critical nuclear receptor, and major regulator in lipid metabolism and inflammation, is suggested to play a protective role against the mitochondrial dysfunction noted in AD. In our study, our established 3D gelatin scaffold model and a well characterized in vivo (APP/PS1) murine model of AD were used to directly investigate the molecular, biochemical and behavioral effects of neuronal stem cell exposure to Aβ to improve understanding of the in vivo etiology of AD. Herein, human neural stem cells (hNSCs) in our 3D model were exposed to Aβ, and had significantly decreased cell viability, which correlated with decreased mRNA and protein expression of LXR, Bcl-2, CREB, PGC1α, NRF-1, and Tfam, and increased caspase 3 and 9 activities. Cotreatment with a synthetic agonist of LXR (TO901317) significantly abrogated these Aβ-mediated effects in hNSCs. Moreover, TO901317 cotreatment both significantly rescues hNSCs from Aβ-mediated decreases in ATP levels and mitochondrial mass, and significantly restores Aβ-induced fragmented mitochondria to almost normal morphology. TO901317 cotreatment also decreases tau aggregates in Aβ-treated hNSCs. Importantly, TO901317 treatment significantly alleviates the impairment of memory, decreases Aβ aggregates and increases proteasome activity in APP/PS1 mice; whereas, these effects were blocked by cotreatment with an LXR antagonist (GSK2033). Together, these novel results improve our mechanistic understanding of the central role of LXR in Aβ-mediated hNSC dysfunction. We also provide preclinical data unveiling the protective effects of using an LXR-dependent agonist, TO901317, to block the toxicity observed in Aβ-exposed hNSCs, which may guide future treatment strategies to slow or prevent neurodegeneration in some AD patients.
Collapse
Affiliation(s)
- Ming-Chang Chiang
- Department of Life Science, College of Science and Engineering, Fu Jen Catholic University, New Taipei City 242, Taiwan.
| | - Christopher J B Nicol
- Departments of Pathology & Molecular Medicine and Biomedical & Molecular Sciences, and Cancer Biology and Genetics Division, Cancer Research Institute, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Shiang-Jiuun Chen
- Department of Life Science and Institute of Ecology and Evolutionary Biology, College of Life Science, National Taiwan University, Taipei 106, Taiwan
| | - Rong-Nan Huang
- Department of Entomology and Research Center for Plant-Medicine, National Taiwan University, Taipei 106, Taiwan
| |
Collapse
|
22
|
Yanguas-Casás N, Torres C, Crespo-Castrillo A, Diaz-Pacheco S, Healy K, Stanton C, Chowen JA, Garcia-Segura LM, Arevalo MA, Cryan JF, de Ceballos ML. High-fat diet alters stress behavior, inflammatory parameters and gut microbiota in Tg APP mice in a sex-specific manner. Neurobiol Dis 2021; 159:105495. [PMID: 34478848 DOI: 10.1016/j.nbd.2021.105495] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 08/13/2021] [Accepted: 08/27/2021] [Indexed: 12/12/2022] Open
Abstract
Long-term high-fat diet (HFD) consumption commonly leads to obesity, a major health concern of western societies and a risk factor for Alzheimer's disease (AD). Both conditions present glial activation and inflammation and show sex differences in their incidence, clinical manifestation, and disease course. HFD intake has an important impact on gut microbiota, the bacteria present in the gut, and microbiota dysbiosis is associated with inflammation and certain mental disorders such as anxiety. In this study, we have analyzed the effects of a prolonged (18 weeks, starting at 7 months of age) HFD on male and female mice, both wild type (WT) and TgAPP mice, a model for AD, investigating the behavioral profile, gut microbiota composition and inflammatory/phagocytosis-related gene expression in hippocampus. In the open-field test, no overt differences in motor activity were observed between male and female or WT and TgAPP mice on a low-fat diet (LFD). However, HFD induced anxiety, as judged by decreased motor activity and increased time in the margins in the open-field, and a trend towards increased immobility time in the tail suspension test, with increased defecation. Intriguingly, female TgAPP mice on HFD showed less immobility and defecation compared to female WT mice on HFD. HFD induced dysbiosis of gut microbiota, resulting in reduced microbiota diversity and abundance compared with LFD fed mice, with some significant differences due to sex and little effect of genotype. Gene expression of pro-inflammatory/phagocytic markers in the hippocampus were not different between male and female WT mice, and in TgAPP mice of both sexes, some cytokines (IL-6 and IFNγ) were higher than in WT mice on LFD, more so in female TgAPP (IL-6). HFD induced few alterations in mRNA expression of inflammatory/phagocytosis-related genes in male mice, whether WT (IL-1β, MHCII), or TgAPP (IL-6). However, in female TgAPP, altered gene expression returned towards control levels following prolonged HFD (IL-6, IL-12β, TNFα, CD36, IRAK4, PYRY6). In summary, we demonstrate that HFD induces anxiogenic symptoms, marked alterations in gut microbiota, and increased expression of inflammatory genes, except for female TgAPP that appear to be resistant to the diet effects. Lifestyle interventions should be introduced to prevent AD onset or exacerbation by reducing inflammation and its associated symptoms; however, our results suggest that the eventual goal of developing prevention and treatment strategies should take sex into consideration.
Collapse
Affiliation(s)
- Natalia Yanguas-Casás
- Cajal Institute, CSIC, 28002 Madrid, Spain; Centre for Biomedical Network Research for Frailty and Healthy Ageing (CIBERFES) Instituto de Salud Carlos III, Madrid, Spain; Lymphoma Research Group, Medical Oncology Department, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana, Majadahonda, Madrid, Spain
| | - Cristina Torres
- Dept Anatomy & Neuroscience, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland; Universitat Rovira i Virgili, Biochemistry and Biotechnology Department, 43007 Tarragona, Spain
| | | | | | - Kiera Healy
- Dept Anatomy & Neuroscience, University College Cork, Cork, Ireland; Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - Catherine Stanton
- Dept Anatomy & Neuroscience, University College Cork, Cork, Ireland; Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - Julie A Chowen
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación la Princesa, 28009 Madrid, Spain; Centre for Biomedical Network Research for Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain; The Madrid Institute for the advanced study of Food (IMDEA de Alimentación), Madrid, Spain
| | - Luis M Garcia-Segura
- Cajal Institute, CSIC, 28002 Madrid, Spain; Centre for Biomedical Network Research for Frailty and Healthy Ageing (CIBERFES) Instituto de Salud Carlos III, Madrid, Spain
| | - Maria Angeles Arevalo
- Cajal Institute, CSIC, 28002 Madrid, Spain; Centre for Biomedical Network Research for Frailty and Healthy Ageing (CIBERFES) Instituto de Salud Carlos III, Madrid, Spain
| | - John F Cryan
- Dept Anatomy & Neuroscience, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | | |
Collapse
|
23
|
Willems S, Zaienne D, Merk D. Targeting Nuclear Receptors in Neurodegeneration and Neuroinflammation. J Med Chem 2021; 64:9592-9638. [PMID: 34251209 DOI: 10.1021/acs.jmedchem.1c00186] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nuclear receptors, also known as ligand-activated transcription factors, regulate gene expression upon ligand signals and present as attractive therapeutic targets especially in chronic diseases. Despite the therapeutic relevance of some nuclear receptors in various pathologies, their potential in neurodegeneration and neuroinflammation is insufficiently established. This perspective gathers preclinical and clinical data for a potential role of individual nuclear receptors as future targets in Alzheimer's disease, Parkinson's disease, and multiple sclerosis, and concomitantly evaluates the level of medicinal chemistry targeting these proteins. Considerable evidence suggests the high promise of ligand-activated transcription factors to counteract neurodegenerative diseases with a particularly high potential of several orphan nuclear receptors. However, potent tools are lacking for orphan receptors, and limited central nervous system exposure or insufficient selectivity also compromises the suitability of well-studied nuclear receptor ligands for functional studies. Medicinal chemistry efforts are needed to develop dedicated high-quality tool compounds for the therapeutic validation of nuclear receptors in neurodegenerative pathologies.
Collapse
Affiliation(s)
- Sabine Willems
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Strasse 9, 60438 Frankfurt, Germany
| | - Daniel Zaienne
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Strasse 9, 60438 Frankfurt, Germany
| | - Daniel Merk
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Strasse 9, 60438 Frankfurt, Germany
| |
Collapse
|
24
|
Fitz NF, Nam KN, Wolfe CM, Letronne F, Playso BE, Iordanova BE, Kozai TDY, Biedrzycki RJ, Kagan VE, Tyurina YY, Han X, Lefterov I, Koldamova R. Phospholipids of APOE lipoproteins activate microglia in an isoform-specific manner in preclinical models of Alzheimer's disease. Nat Commun 2021; 12:3416. [PMID: 34099706 PMCID: PMC8184801 DOI: 10.1038/s41467-021-23762-0] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 05/14/2021] [Indexed: 12/12/2022] Open
Abstract
APOE and Trem2 are major genetic risk factors for Alzheimer's disease (AD), but how they affect microglia response to Aβ remains unclear. Here we report an APOE isoform-specific phospholipid signature with correlation between human APOEε3/3 and APOEε4/4 AD brain and lipoproteins from astrocyte conditioned media of APOE3 and APOE4 mice. Using preclinical AD mouse models, we show that APOE3 lipoproteins, unlike APOE4, induce faster microglial migration towards injected Aβ, facilitate Aβ uptake, and ameliorate Aβ effects on cognition. Bulk and single-cell RNA-seq demonstrate that, compared to APOE4, cortical infusion of APOE3 lipoproteins upregulates a higher proportion of genes linked to an activated microglia response, and this trend is augmented by TREM2 deficiency. In vitro, lack of TREM2 decreases Aβ uptake by APOE4-treated microglia only, suggesting TREM2-APOE interaction. Our study elucidates phenotypic and transcriptional differences in microglial response to Aβ mediated by APOE3 or APOE4 lipoproteins in preclinical models of AD.
Collapse
Affiliation(s)
- Nicholas F Fitz
- Deparment of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kyong Nyon Nam
- Deparment of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Cody M Wolfe
- Deparment of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Florent Letronne
- Deparment of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Brittany E Playso
- Deparment of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Bistra E Iordanova
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Takashi D Y Kozai
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Richard J Biedrzycki
- Deparment of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Valerian E Kagan
- Deparment of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yulia Y Tyurina
- Deparment of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xianlin Han
- Barshop Institute for Longevity and Aging Studies, San Antonio, TX, USA
| | - Iliya Lefterov
- Deparment of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Radosveta Koldamova
- Deparment of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
25
|
Vidal V, Puente A, García-Cerro S, García Unzueta MT, Rueda N, Riancho J, Martínez-Cué C. Bexarotene Impairs Cognition and Produces Hypothyroidism in a Mouse Model of Down Syndrome and Alzheimer's Disease. Front Pharmacol 2021; 12:613211. [PMID: 33935706 PMCID: PMC8082148 DOI: 10.3389/fphar.2021.613211] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 03/15/2021] [Indexed: 12/26/2022] Open
Abstract
All individuals with Down syndrome (DS) eventually develop Alzheimer's disease (AD) neuropathology, including neurodegeneration, increases in β-amyloid (Aβ) expression, and aggregation and neurofibrillary tangles, between the third and fourth decade of their lives. There is currently no effective treatment to prevent AD neuropathology and the associated cognitive degeneration in DS patients. Due to evidence that the accumulation of Aβ aggregates in the brain produces the neurodegenerative cascade characteristic of AD, many strategies which promote the clearance of Aβ peptides have been assessed as potential therapeutics for this disease. Bexarotene, a member of a subclass of retinoids that selectively activates retinoid receptors, modulates several pathways essential for cognitive performance and Aβ clearance. Consequently, bexarotene might be a good candidate to treat AD-associated neuropathology. However, the effects of bexarotene treatment in AD remain controversial. In the present study, we aimed to elucidate whether chronic bexarotene treatment administered to the most commonly used murine model of DS, the Ts65Dn (TS) mouse could reduce Aβ expression in their brains and improve their cognitive abilities. Chronic administration of bexarotene to aged TS mice and their CO littermates for 9 weeks diminished the reference, working, and spatial learning and memory of TS mice, and the spatial memory of CO mice in the Morris water maze. This treatment also produced marked hypoactivity in the plus maze, open field, and hole board tests in TS mice, and in the open field and hole board tests in CO mice. Administration of bexarotene reduced the expression of Aβ1-40, but not of Aβ1-42, in the hippocampi of TS mice. Finally, bexarotene increased Thyroid-stimulating hormone levels in TS mice and reduced Thyroid-stimulating hormone levels in CO mice, while animals of both karyotypes displayed reduced thyroxine levels after bexarotene administration. The bexarotene-induced hypothyroidism could be responsible for the hypoactivity of TS and CO mice and their diminished performance in the Morris water maze. Together, these results do not provide support for the use of bexarotene as a potential treatment of AD neuropathology in the DS population.
Collapse
Affiliation(s)
- Verónica Vidal
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | - Alba Puente
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | - Susana García-Cerro
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain.,CIBERSAM, Madrid, Spain
| | | | - Noemí Rueda
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | - Javier Riancho
- Neurology Service, Hospital Sierrallana-IDIVAL, Torrelavega, Spain.,Department of Medicine and Psychiatry, University of Cantabria, Santander, Spain.,CIBERNED, Madrid, Spain
| | - Carmen Martínez-Cué
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| |
Collapse
|
26
|
Ben Aissa M, Lewandowski CT, Ratia KM, Lee SH, Layden BT, LaDu MJ, Thatcher GRJ. Discovery of Nonlipogenic ABCA1 Inducing Compounds with Potential in Alzheimer's Disease and Type 2 Diabetes. ACS Pharmacol Transl Sci 2021; 4:143-154. [PMID: 33615168 PMCID: PMC7887740 DOI: 10.1021/acsptsci.0c00149] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Indexed: 02/07/2023]
Abstract
Selective liver X receptor (LXR) agonists have been extensively pursued as therapeutics for Alzheimer's disease and related dementia (ADRD) and, for comorbidities such as type 2 diabetes (T2D) and cerebrovascular disease (CVD), disorders with underlying impaired insulin signaling, glucose metabolism, and cholesterol mobilization. The failure of the LXR-focused approach led us to pursue a novel strategy to discover nonlipogenic ATP-binding cassette transporter A1 (ABCA1) inducers (NLAIs): screening for ABCA1-luciferase activation in astrocytoma cells and counterscreening against lipogenic gene upregulation in hepatocarcinoma cells. Beneficial effects of LXRβ agonists mediated by ABCA1 include the following: control of cholesterol and phospholipid efflux to lipid-poor apolipoproteins forming beneficial peripheral HDL and HDL-like particles in the brain and attenuation of inflammation. While rare, ABCA1 variants reduce plasma HDL and correlate with an increased risk of ADRD and CVD. In secondary assays, NLAI hits enhanced cholesterol mobilization and positively impacted in vitro biomarkers associated with insulin signaling, inflammatory response, and biogenic properties. In vivo target engagement was demonstrated after oral administration of NLAIs in (i) mice fed a high-fat diet, a model for obesity-linked T2D, (ii) mice administered LPS, and (iii) mice with accelerated oxidative stress. The lack of adverse effects on lipogenesis and positive effects on multiple biomarkers associated with T2D and ADRD supports this novel phenotypic approach to NLAIs as a platform for T2D and ADRD drug discovery.
Collapse
Affiliation(s)
- Manel Ben Aissa
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago (UIC), Chicago, Illinois 60612, United States
- UICentre
(Drug Discovery @ UIC), University of Illinois
at Chicago (UIC), Chicago, Illinois 60612, United States
| | - Cutler T. Lewandowski
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago (UIC), Chicago, Illinois 60612, United States
| | - Kiira M. Ratia
- HTS
Screening Facility, Research Resources Center, University of Illinois at Chicago (UIC), Chicago, Illinois 60612, United States
| | - Sue H. Lee
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago (UIC), Chicago, Illinois 60612, United States
| | - Brian T. Layden
- Department
of Medicine, University of Illinois at Chicago
(UIC), Chicago, Illinois 60612, United States
| | - Mary Jo LaDu
- Department
of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago (UIC), Chicago, Illinois 60612, United States
| | - Gregory R. J. Thatcher
- Department
of Pharmacology & Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona 85721, United States
| |
Collapse
|
27
|
Chen Y, Strickland MR, Soranno A, Holtzman DM. Apolipoprotein E: Structural Insights and Links to Alzheimer Disease Pathogenesis. Neuron 2020; 109:205-221. [PMID: 33176118 DOI: 10.1016/j.neuron.2020.10.008] [Citation(s) in RCA: 136] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 01/02/2023]
Abstract
Apolipoprotein E (ApoE) is of great interest due to its role as a cholesterol/lipid transporter in the central nervous system (CNS) and as the most influential genetic risk factor for Alzheimer disease (AD). Work over the last four decades has given us important insights into the structure of ApoE and how this might impact the neuropathology and pathogenesis of AD. In this review, we highlight the history and progress in the structural and molecular understanding of ApoE and discuss how these studies on ApoE have illuminated the physiology of ApoE, receptor binding, and interaction with amyloid-β (Aβ). We also identify future areas of study needed to advance our understanding of how ApoE influences neurodegeneration.
Collapse
Affiliation(s)
- Yun Chen
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA; Hope Center for Neurological Disorders, Washington University in St. Louis, St. Louis, MO, USA; Knight Alzheimer's Disease Research Center, Washington University in St. Louis, St. Louis, MO, USA; The Division of Biology and Biomedical Sciences, Washington University in St. Louis, St. Louis, MO, USA; Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO, USA
| | - Michael R Strickland
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA; Hope Center for Neurological Disorders, Washington University in St. Louis, St. Louis, MO, USA; Knight Alzheimer's Disease Research Center, Washington University in St. Louis, St. Louis, MO, USA; The Division of Biology and Biomedical Sciences, Washington University in St. Louis, St. Louis, MO, USA
| | - Andrea Soranno
- Department of Biochemistry and Molecular Biophysics, Washington University in St. Louis, St. Louis, MO, USA; Center for Science & Engineering of Living Systems, Washington University in St. Louis, St. Louis, MO, USA
| | - David M Holtzman
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA; Center for Science & Engineering of Living Systems, Washington University in St. Louis, St. Louis, MO, USA; Knight Alzheimer's Disease Research Center, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
28
|
Pedrini S, Chatterjee P, Hone E, Martins RN. High‐density lipoprotein‐related cholesterol metabolism in Alzheimer’s disease. J Neurochem 2020; 159:343-377. [DOI: 10.1111/jnc.15170] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 08/18/2020] [Accepted: 08/20/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Steve Pedrini
- Sarich Neurosciences Research InstituteEdith Cowan University Nedlands WA Australia
| | - Pratishtha Chatterjee
- Sarich Neurosciences Research InstituteEdith Cowan University Nedlands WA Australia
- Department of Biomedical Sciences Faculty of Medicine, Health and Human Sciences Macquarie University Sydney NSW Australia
| | - Eugene Hone
- Sarich Neurosciences Research InstituteEdith Cowan University Nedlands WA Australia
| | - Ralph N. Martins
- Sarich Neurosciences Research InstituteEdith Cowan University Nedlands WA Australia
- Department of Biomedical Sciences Faculty of Medicine, Health and Human Sciences Macquarie University Sydney NSW Australia
- School of Psychiatry and Clinical Neurosciences University of Western Australia Nedlands WA Australia
| |
Collapse
|
29
|
Reilly AM, Tsai AP, Lin PB, Ericsson AC, Oblak AL, Ren H. Metabolic Defects Caused by High-Fat Diet Modify Disease Risk through Inflammatory and Amyloidogenic Pathways in a Mouse Model of Alzheimer's Disease. Nutrients 2020; 12:nu12102977. [PMID: 33003412 PMCID: PMC7600118 DOI: 10.3390/nu12102977] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/24/2020] [Accepted: 09/25/2020] [Indexed: 12/12/2022] Open
Abstract
High-fat diet (HFD) has been shown to accelerate Alzheimer’s disease (AD) pathology, but the exact molecular and cellular mechanisms remain incompletely understood. Moreover, it is unknown whether AD mice are more susceptible to HFD-induced metabolic dysfunctions. To address these questions, we used 5xFAD mice as an Alzheimer’s disease model to study the physiological and molecular underpinning between HFD-induced metabolic defects and AD pathology. We systematically profiled the metabolic parameters, the gut microbiome composition, and hippocampal gene expression in 5xFAD and wild type (WT) mice fed normal chow diet and HFD. HFD feeding impaired energy metabolism in male 5xFAD mice, leading to increased locomotor activity, energy expenditure, and food intake. 5xFAD mice on HFD had elevated circulating lipids and worsened glucose intolerance. HFD caused profound changes in gut microbiome compositions, though no difference between genotype was detected. We measured hippocampal mRNAs related to AD neuropathology and neuroinflammation and showed that HFD elevated the expression of apoptotic, microglial, and amyloidogenic genes in 5xFAD mice. Pathway analysis revealed that differentially regulated genes were involved in insulin signaling, cytokine signaling, cellular stress, and neurotransmission. Collectively, our results showed that 5xFAD mice were more susceptible to HFD-induced metabolic dysregulation and suggest that targeting metabolic dysfunctions can ameliorate AD symptoms via effects on insulin signaling and neuroinflammation in the hippocampus.
Collapse
Affiliation(s)
- Austin M. Reilly
- Stark Neurosciences Research Institute, Medical Neuroscience Graduate Program, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.M.R.); (A.P.T.); (P.B.L.); (A.L.O.)
| | - Andy P. Tsai
- Stark Neurosciences Research Institute, Medical Neuroscience Graduate Program, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.M.R.); (A.P.T.); (P.B.L.); (A.L.O.)
| | - Peter B. Lin
- Stark Neurosciences Research Institute, Medical Neuroscience Graduate Program, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.M.R.); (A.P.T.); (P.B.L.); (A.L.O.)
| | - Aaron C. Ericsson
- Metagenomics Center, University of Missouri, Columbia, MO 65201, USA;
| | - Adrian L. Oblak
- Stark Neurosciences Research Institute, Medical Neuroscience Graduate Program, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.M.R.); (A.P.T.); (P.B.L.); (A.L.O.)
| | - Hongxia Ren
- Stark Neurosciences Research Institute, Medical Neuroscience Graduate Program, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (A.M.R.); (A.P.T.); (P.B.L.); (A.L.O.)
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Correspondence: ; Tel.: +1-317-274-1567
| |
Collapse
|
30
|
Fitz NF, Wolfe CM, Playso BE, Biedrzycki RJ, Lu Y, Nam KN, Lefterov I, Koldamova R. Trem2 deficiency differentially affects phenotype and transcriptome of human APOE3 and APOE4 mice. Mol Neurodegener 2020; 15:41. [PMID: 32703241 PMCID: PMC7379780 DOI: 10.1186/s13024-020-00394-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 07/16/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Alzheimer's Disease (AD) is a neurodegenerative disorder influenced by aging and genetic risk factors. The inheritance of APOEε4 and variants of Triggering Receptor Expressed on Myeloid cells 2 (TREM2) are major genetic risk factors for AD. Recent studies showed that APOE binds to TREM2, thus raising the possibility of an APOE-TREM2 interaction that can modulate AD pathology. METHODS The aim of this study was to investigate this interaction using complex AD model mice - a crossbreed of Trem2ko and APP/PSEN1dE9 mice expressing human APOE3 or APOE4 isoforms (APP/E3 and APP/E4 respectively), and their WT littermates (E3 and E4), and evaluate cognition, steady-state amyloid load, plaque compaction, plaque growth rate, glial response, and brain transcriptome. RESULTS In both, APP/E3 and APP/E4 mice, Trem2 deletion reduced plaque compaction but did not significantly affect steady-state plaque load. Importantly, the lack of TREM2 increased plaque growth that negatively correlated to the diminished microglia barrier, an effect most pronounced at earlier stages of amyloid deposition. We also found that Trem2 deficiency significantly decreased plaque-associated APOE protein in APP/E4 but not in APP/E3 mice in agreement with RNA-seq data. Interestingly, we observed a significant decrease of Apoe mRNA expression in plaque-associated microglia of APP/E4/Trem2ko vs APP/E4 mice. The absence of TREM2, worsened cognitive performance in APP transgenic mice but not their WT littermates. Gene expression analysis identified Trem2 signature - a cluster of highly connected immune response genes, commonly downregulated as a result of Trem2 deletion in all genotypes including APP and WT littermates. Furthermore, we identified sets of genes that were affected in TREM2- and APOE isoform-dependent manner. Among them were Clec7a and Csf1r upregulated in APP/E4 vs APP/E3 mice, a result further validated by in situ hybridization analysis. In contrast, Tyrobp and several genes involved in the C1Q complement cascade had a higher expression level in APP/E3 versus their APP/E4 counterparts. CONCLUSIONS Our data demonstrate that lack of Trem2 differentially impacts the phenotype and brain transcriptome of APP mice expressing human APOE isoforms. The changes probably reflect the different effect of APOE isoforms on amyloid deposition.
Collapse
Affiliation(s)
- Nicholas F. Fitz
- Department of Environmental & Occupational Health, University of Pittsburgh, 130 De Soto Street, Pittsburgh, PA 15261 USA
| | - Cody M. Wolfe
- Department of Environmental & Occupational Health, University of Pittsburgh, 130 De Soto Street, Pittsburgh, PA 15261 USA
| | - Brittany E. Playso
- Department of Environmental & Occupational Health, University of Pittsburgh, 130 De Soto Street, Pittsburgh, PA 15261 USA
| | - Richard J. Biedrzycki
- Department of Environmental & Occupational Health, University of Pittsburgh, 130 De Soto Street, Pittsburgh, PA 15261 USA
| | - Yi Lu
- Department of Environmental & Occupational Health, University of Pittsburgh, 130 De Soto Street, Pittsburgh, PA 15261 USA
| | - Kyong Nyon Nam
- Department of Environmental & Occupational Health, University of Pittsburgh, 130 De Soto Street, Pittsburgh, PA 15261 USA
| | - Iliya Lefterov
- Department of Environmental & Occupational Health, University of Pittsburgh, 130 De Soto Street, Pittsburgh, PA 15261 USA
| | - Radosveta Koldamova
- Department of Environmental & Occupational Health, University of Pittsburgh, 130 De Soto Street, Pittsburgh, PA 15261 USA
| |
Collapse
|
31
|
Lewandowski CT, Maldonado Weng J, LaDu MJ. Alzheimer's disease pathology in APOE transgenic mouse models: The Who, What, When, Where, Why, and How. Neurobiol Dis 2020; 139:104811. [PMID: 32087290 DOI: 10.1016/j.nbd.2020.104811] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 02/01/2020] [Accepted: 02/18/2020] [Indexed: 02/07/2023] Open
Abstract
The focus on amyloid plaques and neurofibrillary tangles has yielded no Alzheimer's disease (AD) modifying treatments in the past several decades, despite successful studies in preclinical mouse models. This inconsistency has caused a renewed focus on improving the fidelity and reliability of AD mouse models, with disparate views on how this improvement can be accomplished. However, the interactive effects of the universal biological variables of AD, which include age, APOE genotype, and sex, are often overlooked. Age is the greatest risk factor for AD, while the ε4 allele of the human APOE gene, encoding apolipoprotein E, is the greatest genetic risk factor. Sex is the final universal biological variable of AD, as females develop AD at almost twice the rate of males and, importantly, female sex exacerbates the effects of APOE4 on AD risk and rate of cognitive decline. Therefore, this review evaluates the importance of context for understanding the role of APOE in preclinical mouse models. Specifically, we detail how human AD pathology is mirrored in current transgenic mouse models ("What") and describe the critical need for introducing human APOE into these mouse models ("Who"). We next outline different methods for introducing human APOE into mice ("How") and highlight efforts to develop temporally defined and location-specific human apoE expression models ("When" and "Where"). We conclude with the importance of choosing the human APOE mouse model relevant to the question being addressed, using the selection of transgenic models for testing apoE-targeted therapeutics as an example ("Why").
Collapse
Affiliation(s)
- Cutler T Lewandowski
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, 833 S. Wood St., Chicago, IL 60612, USA.
| | - Juan Maldonado Weng
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, 808 S. Wood St., Chicago, IL 60612, USA.
| | - Mary Jo LaDu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, 808 S. Wood St., Chicago, IL 60612, USA.
| |
Collapse
|
32
|
Hahm JR, Jo MH, Ullah R, Kim MW, Kim MO. Metabolic Stress Alters Antioxidant Systems, Suppresses the Adiponectin Receptor 1 and Induces Alzheimer's Like Pathology in Mice Brain. Cells 2020; 9:cells9010249. [PMID: 31963819 PMCID: PMC7016950 DOI: 10.3390/cells9010249] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 01/09/2020] [Accepted: 01/15/2020] [Indexed: 01/02/2023] Open
Abstract
Oxidative stress and insulin resistance play major roles in numerous neurodegenerative diseases, including Alzheimer’s disease (AD). A high-fat diet induces obesity-associated oxidative stress, neuronal insulin resistance, microglial activation, and neuroinflammation, which are considered important risk factors for neurodegeneration. Obesity-related metabolic dysfunction is a risk factor for cognitive decline. The present study aimed to elucidate whether chronic consumption of a high-fat diet (HFD; 24 weeks) can induce insulin resistance, neuroinflammation, and amyloid beta (Aβ) deposition in mouse brains. Male C57BL/6N mice were used for a high-fat diet (HFD)-induced pre-clinical model of obesity. The protein expression levels were examined via Western blot, immunofluorescence, and the behavior analysis was performed using the Morris water maze test. To obtain metabolic parameters, insulin sensitivity and glucose tolerance tests were performed. We found that metabolic perturbations from the chronic consumption of HFD elevated neuronal oxidative stress and insulin resistance through adiponectin receptor (AdipoR1) suppression in HFD-fed mice. Similarly, our in vitro results also indicated that knockdown of AdipoR1 in the embryonic mouse hippocampal cell line mHippoE-14 leads to increased oxidative stress in neurons. In addition, HFD markedly increased neuroinflammatory markers’ glial activation in the cortex and hippocampus regions of HFD mouse brains. More importantly, we observed that AdipoR1 suppression increased the amyloidogenic pathway both in vivo and in vitro. Furthermore, deregulated synaptic proteins and behavioral deficits were observed in the HFD mouse brains. Taken together, our findings suggest that excessive consumption of an HFD has a profound impact on brain function, which involves the acceleration of cognitive impairment due to increased obesity-associated oxidative stress, insulin resistance, and neuroinflammation, which ultimately may cause early onset of Alzheimer’s pathology via the suppression of AdipoR1 signaling in the brain.
Collapse
Affiliation(s)
- Jong Ryeal Hahm
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Gyeongsang National University Hospital and Institute of Health Sciences and Department of Internal Medicine, College of Medicine, Gyeongsang National University, Jinju 52828, Korea;
| | - Myeung Hoon Jo
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (M.H.J.); (R.U.); (M.W.K.)
| | - Rahat Ullah
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (M.H.J.); (R.U.); (M.W.K.)
| | - Min Woo Kim
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (M.H.J.); (R.U.); (M.W.K.)
| | - Myeong Ok Kim
- Division of Life Sciences and Applied Life Science (BK 21plus), College of Natural Science, Gyeongsang National University, Jinju 52828, Korea; (M.H.J.); (R.U.); (M.W.K.)
- Correspondence: ; Tel.: +82-55-772-1345; Fax: +82-55-772-2656
| |
Collapse
|
33
|
Román G, Jackson R, Reis J, Román A, Toledo J, Toledo E. Extra-virgin olive oil for potential prevention of Alzheimer disease. Rev Neurol (Paris) 2019; 175:705-723. [DOI: 10.1016/j.neurol.2019.07.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 07/12/2019] [Accepted: 07/12/2019] [Indexed: 02/07/2023]
|
34
|
Fitz NF, Nam KN, Koldamova R, Lefterov I. Therapeutic targeting of nuclear receptors, liver X and retinoid X receptors, for Alzheimer's disease. Br J Pharmacol 2019; 176:3599-3610. [PMID: 30924124 PMCID: PMC6715597 DOI: 10.1111/bph.14668] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 02/15/2019] [Accepted: 02/24/2019] [Indexed: 12/18/2022] Open
Abstract
After 15 years of research into Alzheimer's disease (AD) therapeutics, including billions of US dollars provided by federal agencies, pharmaceutical companies, and private foundations, there are still no meaningful therapies that can delay the onset or slow the progression of AD. An understanding of the proteolytic processing of amyloid precursor protein (APP) and the hypothesis that pathogenic mechanisms in familial and sporadic forms of AD are very similar led to the assumption that pharmacological inhibition of secretases or immunological approaches to clear amyloid depositions in the brain would have been the core to drug discovery strategies and successful therapies. However, there are other understudied approaches including targeting genes, gene networks, and metabolic pathways outside the proteolytic processing of APP. The advancement of newly developed sequencing technologies and mass spectrometry, as well as the availability of animal models expressing human apolipoprotein E isoforms, has been critical in rationalizing additional AD therapeutics. The purpose of this review is to present one of those approaches, based on the role of ligand-activated nuclear liver X and retinoid X receptors in the brain. This therapeutic approach was initially proposed utilizing in vitro models 15 years ago and has since been examined in numerous studies using AD-like mouse models. LINKED ARTICLES: This article is part of a themed section on Therapeutics for Dementia and Alzheimer's Disease: New Directions for Precision Medicine. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.18/issuetoc.
Collapse
Affiliation(s)
- Nicholas F Fitz
- Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Kyong Nyon Nam
- Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Radosveta Koldamova
- Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Iliya Lefterov
- Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
35
|
Saint-Pol J, Gosselet F. Oxysterols and the NeuroVascular Unit (NVU): A far true love with bright and dark sides. J Steroid Biochem Mol Biol 2019; 191:105368. [PMID: 31026511 DOI: 10.1016/j.jsbmb.2019.04.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 04/10/2019] [Accepted: 04/22/2019] [Indexed: 12/13/2022]
Abstract
The brain is isolated from the whole body by the blood-brain barrier (BBB) which is located in brain microvessel endothelial cells (ECs). Through physical and metabolic properties induced by brain pericytes, astrocytes and neurons (these cells and the ECs referred to as the neurovascular unit (NVU)), the BBB hardly restricts exchanges of molecules between the brain and the bloodstream. Among them, cholesterol exchanges between these two compartments are very limited and occur through the transport of LDLs across the BBB. Oxysterols (mainly 24S and 27-hydroxycholesterol) daily cross the BBB and regulate molecule/cholesterol exchanges via Liver X nuclear Receptors (LXRs). In addition, these oxysterols have been linked to pathological processes in neurodegenerative diseases such as Alzheimer's disease. Here we propose an overview of the actual knowledge concerning oxysterols and the NVU cells in physiological and in Alzheimer's disease.
Collapse
Affiliation(s)
- Julien Saint-Pol
- University of Artois, Blood-Brain Barrier Laboratory (BBB Lab), EA2465, F-62300 Lens, France.
| | - Fabien Gosselet
- University of Artois, Blood-Brain Barrier Laboratory (BBB Lab), EA2465, F-62300 Lens, France
| |
Collapse
|
36
|
Leyrolle Q, Layé S, Nadjar A. Direct and indirect effects of lipids on microglia function. Neurosci Lett 2019; 708:134348. [PMID: 31238131 DOI: 10.1016/j.neulet.2019.134348] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 06/05/2019] [Accepted: 06/21/2019] [Indexed: 02/07/2023]
Abstract
Microglia are key players in brain function by maintaining brain homeostasis across lifetime. They participate to brain development and maturation through their ability to release neurotrophic factors, to remove immature synapses or unnecessary neural progenitors. They modulate neuronal activity in healthy adult brains and they also orchestrate the neuroinflammatory response in various pathophysiological contexts such as aging and neurodegenerative diseases. One of the main features of microglia is their high sensitivity to environmental factors, partly via the expression of a wide range of receptors. Recent data pinpoint that dietary fatty acids modulate microglia function. Both the quantity and the type of fatty acid are potent modulators of microglia physiology. The present review aims at dissecting the current knowledge on the direct and indirect mechanisms (focus on gut microbiota and hormones) through which fatty acids influence microglial physiology. We summarize main discoveries from in vitro and in vivo models on fatty acid-mediated microglial modulation. All these studies represent a promising field of research that could promote using nutrition as a novel therapeutic or preventive tool in diseases involving microglia dysfunctions.
Collapse
Affiliation(s)
- Q Leyrolle
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Univ. Bordeaux, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France
| | - S Layé
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Univ. Bordeaux, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France
| | - A Nadjar
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Univ. Bordeaux, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France.
| |
Collapse
|
37
|
Cline EN, Bicca MA, Viola KL, Klein WL. The Amyloid-β Oligomer Hypothesis: Beginning of the Third Decade. J Alzheimers Dis 2019; 64:S567-S610. [PMID: 29843241 PMCID: PMC6004937 DOI: 10.3233/jad-179941] [Citation(s) in RCA: 543] [Impact Index Per Article: 108.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The amyloid-β oligomer (AβO) hypothesis was introduced in 1998. It proposed that the brain damage leading to Alzheimer’s disease (AD) was instigated by soluble, ligand-like AβOs. This hypothesis was based on the discovery that fibril-free synthetic preparations of AβOs were potent CNS neurotoxins that rapidly inhibited long-term potentiation and, with time, caused selective nerve cell death (Lambert et al., 1998). The mechanism was attributed to disrupted signaling involving the tyrosine-protein kinase Fyn, mediated by an unknown toxin receptor. Over 4,000 articles concerning AβOs have been published since then, including more than 400 reviews. AβOs have been shown to accumulate in an AD-dependent manner in human and animal model brain tissue and, experimentally, to impair learning and memory and instigate major facets of AD neuropathology, including tau pathology, synapse deterioration and loss, inflammation, and oxidative damage. As reviewed by Hayden and Teplow in 2013, the AβO hypothesis “has all but supplanted the amyloid cascade.” Despite the emerging understanding of the role played by AβOs in AD pathogenesis, AβOs have not yet received the clinical attention given to amyloid plaques, which have been at the core of major attempts at therapeutics and diagnostics but are no longer regarded as the most pathogenic form of Aβ. However, if the momentum of AβO research continues, particularly efforts to elucidate key aspects of structure, a clear path to a successful disease modifying therapy can be envisioned. Ensuring that lessons learned from recent, late-stage clinical failures are applied appropriately throughout therapeutic development will further enable the likelihood of a successful therapy in the near-term.
Collapse
Affiliation(s)
- Erika N Cline
- Department of Neurobiology, Cognitive Neurology and Alzheimer's Disease Center, International Institute for Nanotechnology, and Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| | - Maíra Assunção Bicca
- Department of Neurobiology, Cognitive Neurology and Alzheimer's Disease Center, International Institute for Nanotechnology, and Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| | - Kirsten L Viola
- Department of Neurobiology, Cognitive Neurology and Alzheimer's Disease Center, International Institute for Nanotechnology, and Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| | - William L Klein
- Department of Neurobiology, Cognitive Neurology and Alzheimer's Disease Center, International Institute for Nanotechnology, and Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, USA
| |
Collapse
|
38
|
Wakabayashi T, Yamaguchi K, Matsui K, Sano T, Kubota T, Hashimoto T, Mano A, Yamada K, Matsuo Y, Kubota N, Kadowaki T, Iwatsubo T. Differential effects of diet- and genetically-induced brain insulin resistance on amyloid pathology in a mouse model of Alzheimer's disease. Mol Neurodegener 2019; 14:15. [PMID: 30975165 PMCID: PMC6460655 DOI: 10.1186/s13024-019-0315-7] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 03/15/2019] [Indexed: 12/21/2022] Open
Abstract
Background Based on epidemiological and experimental studies, type 2 diabetes mellitus (T2DM), especially insulin resistance that comprises the core mechanism of T2DM, has been recognized as a significant risk factor for Alzheimer’s disease (AD). Studies in humans and diabetic AD model mice have indicated a correlation between insulin resistance and increased amyloid deposition in the brain. Paradoxically, mice with targeted disruption of genes involved in the insulin signaling pathway showed protective effects against the AD-related pathology. These conflicting observations raise an issue as to the relationship between dysregulation of insulin signaling and AD pathophysiology. Methods To study the causal relations and molecular mechanisms underlying insulin resistance-induced exacerbation of amyloid pathology, we investigated the chronological changes in the development of insulin resistance and amyloid pathology in two independent insulin-resistant AD mouse models, i.e., long-term high-fat diet (HFD) feeding and genetic disruption of Irs2, in combination with dietary interventions. In addition to biochemical and histopathological analyses, we examined the in vivo dynamics of brain amyloid-β (Aβ) and insulin by microdialysis technique. Results HFD-fed diabetic AD model mice displayed a reduced brain response to peripheral insulin stimulation and a decreased brain to plasma ratio of insulin during the hyperinsulinemic clamp. Diet-induced defective insulin action in the brain was accompanied by a decreased clearance of the extracellular Aβ in vivo and an exacerbation of brain amyloid pathology. These noxious effects of the HFD both on insulin sensitivity and on Aβ deposition in brains were reversibly attenuated by dietary interventions. Importantly, HFD feeding accelerated Aβ deposition also in the brains of IRS-2-deficient AD mice. Conclusions Our results suggested a causal and reversible association of brain Aβ metabolism and amyloid pathology by diet-dependent, but not genetically-induced, insulin-resistance. These observations raise the possibility that the causal factors of insulin resistance, e.g., metabolic stress or inflammation induced by HFD feeding, but not impaired insulin signaling per se, might be directly involved in the acceleration of amyloid pathology in the brain. Electronic supplementary material The online version of this article (10.1186/s13024-019-0315-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tomoko Wakabayashi
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,Department of Innovative Dementia Prevention, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Kazuki Yamaguchi
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Kentaro Matsui
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Toshiharu Sano
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Tetsuya Kubota
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Kanagawa, 230-0045, Japan
| | - Tadafumi Hashimoto
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,Department of Innovative Dementia Prevention, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Ayako Mano
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Kaoru Yamada
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Yuko Matsuo
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Naoto Kubota
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan.,Department of Clinical Nutrition Therapy, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Takashi Kadowaki
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan.,Department of Prevention of Diabetes and Lifestyle-Related Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan.,Department of Metabolism and Nutrition, Mizonokuchi Hospital, Faculty of Medicine, Teikyo University, Tokyo, 213-8507, Japan
| | - Takeshi Iwatsubo
- Department of Neuropathology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| |
Collapse
|
39
|
Marwarha G, Claycombe-Larson K, Lund J, Schommer J, Ghribi O. A Diet Enriched in Palmitate and Deficient in Linoleate Exacerbates Oxidative Stress and Amyloid-β Burden in the Hippocampus of 3xTg-AD Mouse Model of Alzheimer’s Disease. J Alzheimers Dis 2019; 68:219-237. [DOI: 10.3233/jad-180835] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Gurdeep Marwarha
- Department of Biomedical Sciences, School of Medicine & Health Sciences, University of North Dakota, Grand Forks, ND, USA
| | - Kate Claycombe-Larson
- U.S. Department of Agriculture, Agricultural Research Service, Grand Forks Human Nutrition Research Center, Grand Forks, ND, USA
| | - Jonah Lund
- Department of Biomedical Sciences, School of Medicine & Health Sciences, University of North Dakota, Grand Forks, ND, USA
| | - Jared Schommer
- Department of Biomedical Sciences, School of Medicine & Health Sciences, University of North Dakota, Grand Forks, ND, USA
| | - Othman Ghribi
- Department of Biomedical Sciences, School of Medicine & Health Sciences, University of North Dakota, Grand Forks, ND, USA
| |
Collapse
|
40
|
Mechanisms Associated with Type 2 Diabetes as a Risk Factor for Alzheimer-Related Pathology. Mol Neurobiol 2019; 56:5815-5834. [PMID: 30684218 DOI: 10.1007/s12035-019-1475-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 01/10/2019] [Indexed: 12/19/2022]
Abstract
Current evidence suggests dementia and pathology in Alzheimer's Disease (AD) are both dependent and independent of amyloid processing and can be induced by multiple 'hits' on vital neuronal functions. Type 2 diabetes (T2D) poses the most important risk factor for developing AD after ageing and dysfunctional IR/PI3K/Akt signalling is a major contributor in both diseases. We developed a model of T2D, coupling subdiabetogenic doses of streptozotocin (STZ) with a human junk food (HJF) diet to more closely mimic the human condition. Over 35 weeks, this induced classic signs of T2D (hyperglycemia and insulin dysfunction) and a modest, but stable deficit in spatial recognition memory, with very little long-term modification of proteins in or associated with IR/PI3K/Akt signalling in CA1 of the hippocampus. Intracerebroventricular infusion of soluble amyloid beta 42 (Aβ42) to mimic the early preclinical rise in Aβ alone induced a more severe, but short-lasting deficits in memory and deregulation of proteins. Infusion of Aβ on the T2D phenotype exacerbated and prolonged the memory deficits over approximately 4 months, and induced more severe aberrant regulation of proteins associated with autophagy, inflammation and glucose uptake from the periphery. A mild form of environmental enrichment transiently rescued memory deficits and could reverse the regulation of some, but not all protein changes. Together, these data identify mechanisms by which T2D could create a modest dysfunctional neuronal milieu via multiple and parallel inputs that permits the development of pathological events identified in AD and memory deficits when Aβ levels are transiently effective in the brain.
Collapse
|
41
|
Cao J, Hou J, Ping J, Cai D. Advances in developing novel therapeutic strategies for Alzheimer's disease. Mol Neurodegener 2018; 13:64. [PMID: 30541602 PMCID: PMC6291983 DOI: 10.1186/s13024-018-0299-8] [Citation(s) in RCA: 156] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 11/28/2018] [Indexed: 12/16/2022] Open
Abstract
Alzheimer's Disease (AD), the most prevalent neurodegenerative disease of aging, affects one in eight older Americans. Nearly all drug treatments tested for AD today have failed to show any efficacy. There is a great need for therapies to prevent and/or slow the progression of AD. The major challenge in AD drug development is lack of clarity about the mechanisms underlying AD pathogenesis and pathophysiology. Several studies support the notion that AD is a multifactorial disease. While there is abundant evidence that amyloid plays a role in AD pathogenesis, other mechanisms have been implicated in AD such as tangle formation and spread, dysregulated protein degradation pathways, neuroinflammation, and loss of support by neurotrophic factors. Therefore, current paradigms of AD drug design have been shifted from single target approach (primarily amyloid-centric) to developing drugs targeted at multiple disease aspects, and from treating AD at later stages of disease progression to focusing on preventive strategies at early stages of disease development. Here, we summarize current strategies and new trends of AD drug development, including pre-clinical and clinical trials that target different aspects of disease (mechanism-based versus non-mechanism based, e.g. symptomatic treatments, lifestyle modifications and risk factor management).
Collapse
Affiliation(s)
- Jiqing Cao
- James J Peters VA Medical Center, Research & Development, Bronx, NY 10468 USA
- Department of Neurology, Alzheimer Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
- The Central Hospital of The Hua Zhong University of Science and Technology, Wuhan, China
| | - Jianwei Hou
- James J Peters VA Medical Center, Research & Development, Bronx, NY 10468 USA
- Department of Neurology, Alzheimer Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Jing Ping
- The Central Hospital of The Hua Zhong University of Science and Technology, Wuhan, China
| | - Dongming Cai
- James J Peters VA Medical Center, Research & Development, Bronx, NY 10468 USA
- Department of Neurology, Alzheimer Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
- The Central Hospital of The Hua Zhong University of Science and Technology, Wuhan, China
| |
Collapse
|
42
|
Montoliu-Gaya L, Güell-Bosch J, Esquerda-Canals G, Roda AR, Serra-Mir G, Lope-Piedrafita S, Sánchez-Quesada JL, Villegas S. Differential effects of apoE and apoJ mimetic peptides on the action of an anti-Aβ scFv in 3xTg-AD mice. Biochem Pharmacol 2018; 155:380-392. [PMID: 30026023 DOI: 10.1016/j.bcp.2018.07.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 07/13/2018] [Indexed: 12/31/2022]
Abstract
Anti-Aβ immunotherapy has emerged as a promising approach to treat Alzheimer's disease (AD). The single-chain variable fragment scFv-h3D6 is an anti-Aβ antibody fragment that lacks the Fc region, which is associated with the induction of microglial reactivity by the full-length monoclonal antibody bapineuzumab. ScFv-h3D6 was previously shown to restore the levels of apolipoprotein E (apoE) and apolipoprotein J (apoJ) in a triple-transgenic-AD (3xTg-AD) mouse model. Since apoE and apoJ play an important role in the development of AD, we aimed to study the in vivo effect of the combined therapy of scFv-h3D6 with apoE and apoJ mimetic peptides (MPs). Four-and-a-half-month-old 3xTg-AD mice were treated for six weeks with scFv-h3D6, apoE-MP, apoJ-MP, or a combination of scFv-h3D6 with each of the MPs, or a vehicle, and then the results were compared to non-transgenic mice. Magnetic Resonance Imaging showed a general tendency of the different treatments to protect against the reduction in brain volume. Aβ burden decreased after treatment with scFv-h3D6, apoE-MP, or apoJ-MP, but the effect was not as evident with the combined therapies. In terms of glial reactivity, apoE-MP showed a potent anti-inflammatory effect that was eased by the presence of scFv-h3D6, whereas the combination of apoJ-MP and scFv-h3D6 was not detrimental. ScFv-h3D6 alone did not induce microglial reactivity, as full-length antibodies do; rather, it reduced it. Endogenous apoE and apoJ levels were decreased by scFv-h3D6, but the MPs lead to a simultaneous increase of both apolipoproteins. While apoE-MP and apoJ-MP demonstrated different effects in the combined therapies with scFv-h3D6, they did not improve the overall protective effect of scFv-h3D6 in reducing the Aβ burden, apolipoproteins levels or microglial reactivity.
Collapse
Affiliation(s)
- Laia Montoliu-Gaya
- Protein Folding and Stability Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain.
| | - Jofre Güell-Bosch
- Protein Folding and Stability Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain.
| | - Gisela Esquerda-Canals
- Protein Folding and Stability Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain.
| | - Alejandro R Roda
- Protein Folding and Stability Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain.
| | - Gabriel Serra-Mir
- Protein Folding and Stability Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain.
| | - Silvia Lope-Piedrafita
- Servei de Ressonància Magnètica Nuclear, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain; Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 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 Metabolic Diseases (CIBERDEM), Spain.
| | - Sandra Villegas
- Protein Folding and Stability Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Barcelona, Spain.
| |
Collapse
|
43
|
Zhou MM, Ding L, Wen M, Che HX, Huang JQ, Zhang TT, Xue CH, Mao XZ, Wang YM. Mechanisms of DHA-enriched phospholipids in improving cognitive deficits in aged SAMP8 mice with high-fat diet. J Nutr Biochem 2018; 59:64-75. [PMID: 29986309 DOI: 10.1016/j.jnutbio.2018.05.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Revised: 04/15/2018] [Accepted: 05/16/2018] [Indexed: 01/29/2023]
Abstract
Recent studies have shown that a high-fat diet (HFD) is involved in both metabolic dysfunction and cognitive deficiency and that docosahexaenoic-acid-enriched phospholipids (DHA-PLs) have beneficial effects on obesity and cognitive impairment. However, there are only a few studies comparing differences between DHA-PC and DHA-PS in HFD-induced Alzheimer's disease (AD) models. After 8 weeks feeding with HFD, 10-month-old SAMP8 mice were fed with 1% (w/w) DHA-PC or 1% DHA-PS (biosynthesized from DHA-PC) for 8 weeks; we then tested the behavioral performances in the Barnes maze test and Morris maze test. The changes of the generation and accumulation of Aβ, oxidative stress, apoptosis, neuroinflammation and neurotrophic factors were also measured. The results indicated that both DHA-PC and DHA-PS significantly improved the metabolic disorders and cognitive deficits. Both DHA-PC and DHA-PS could ameliorate oxidative stress, and DHA-PS presented more notable benefits than DHA-PC on Aβ pathology, mitochondrial damage, neuroinflammation and neurotrophic factors; DHA-PS was for the first time found to increase the production of insoluble Aβ (less pathogenic) in this AD model. These data suggest that DHA-PLs can significantly improve cognitive deficiency, and the molecular mechanisms for this closely relate to the phospholipid polar groups.
Collapse
Affiliation(s)
- Miao-Miao Zhou
- College of Food Science and Engineering, Ocean University of China, No. 5 Yushan Road, Qingdao, Shandong Province 266003, PR China
| | - Lin Ding
- College of Food Science and Engineering, Ocean University of China, No. 5 Yushan Road, Qingdao, Shandong Province 266003, PR China
| | - Min Wen
- College of Food Science and Engineering, Ocean University of China, No. 5 Yushan Road, Qingdao, Shandong Province 266003, PR China
| | - Hong-Xia Che
- College of Food Science and Engineering, Ocean University of China, No. 5 Yushan Road, Qingdao, Shandong Province 266003, PR China
| | - Jia-Qi Huang
- College of Food Science and Engineering, Ocean University of China, No. 5 Yushan Road, Qingdao, Shandong Province 266003, PR China
| | - Tian-Tian Zhang
- College of Food Science and Engineering, Ocean University of China, No. 5 Yushan Road, Qingdao, Shandong Province 266003, PR China
| | - Chang-Hu Xue
- College of Food Science and Engineering, Ocean University of China, No. 5 Yushan Road, Qingdao, Shandong Province 266003, PR China; Qingdao National Laboratory for Marine Science and Technology, Laboratory of Marine Drugs & Biological products, Qingdao 266237, PR China
| | - Xiang-Zhao Mao
- College of Food Science and Engineering, Ocean University of China, No. 5 Yushan Road, Qingdao, Shandong Province 266003, PR China; Qingdao National Laboratory for Marine Science and Technology, Laboratory of Marine Drugs & Biological products, Qingdao 266237, PR China.
| | - Yu-Ming Wang
- College of Food Science and Engineering, Ocean University of China, No. 5 Yushan Road, Qingdao, Shandong Province 266003, PR China; Qingdao National Laboratory for Marine Science and Technology, Laboratory of Marine Drugs & Biological products, Qingdao 266237, PR China.
| |
Collapse
|
44
|
Chen M, Yang F, Kang J, Gan H, Yang X, Lai X, Gao Y. Identfication of Potent LXRβ-Selective Agonists without LXRα Activation by In Silico Approaches. Molecules 2018; 23:molecules23061349. [PMID: 29867043 PMCID: PMC6099648 DOI: 10.3390/molecules23061349] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Revised: 05/30/2018] [Accepted: 05/31/2018] [Indexed: 11/16/2022] Open
Abstract
Activating Liver X receptors (LXRs) represents a promising therapeutic option for dyslipidemia. However, activating LXRα may cause undesired lipogenic effects. Discovery of highly LXRβ-selective agonists without LXRα activation were indispensable for dyslipidemia. In this study, in silico approaches were applied to develop highly potent LXRβ-selective agonists based on a series of newly reported 3-(4-(2-propylphenoxy)butyl)imidazolidine-2,4-dione-based LXRα/β dual agonists. Initially, Kohonen and stepwise multiple linear regression SW-MLR were performed to construct models for LXRβ agonists and LXRα agonists based on the structural characteristics of LXRα/β dual agonists, respectively. The obtained LXRβ agonist model gave a good predictive ability (R2train = 0.837, R2test = 0.843, Q2LOO = 0.715), and the LXRα agonist model produced even better predictive ability (R2train = 0.968, R2test = 0.914, Q2LOO = 0.895). Also, the two QSAR models were independent and can well distinguish LXRβ and LXRα activity. Then, compounds in the ZINC database met the lower limit of structural similarity of 0.7, compared to the 3-(4-(2-propylphenoxy)butyl)imidazolidine-2,4-dione scaffold subjected to our QSAR models, which resulted in the discovery of ZINC55084484 with an LXRβ prediction value of pEC50 equal to 7.343 and LXRα prediction value of pEC50 equal to −1.901. Consequently, nine newly designed compounds were proposed as highly LXRβ-selective agonists based on ZINC55084484 and molecular docking, of which LXRβ prediction values almost exceeded 8 and LXRα prediction values were below 0.
Collapse
Affiliation(s)
- Meimei Chen
- College of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China.
- College of Chemistry and Materials Science, Fujian Normal University, Fuzhou 350007, China.
| | - Fafu Yang
- College of Chemistry and Materials Science, Fujian Normal University, Fuzhou 350007, China.
| | - Jie Kang
- College of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China.
| | - Huijuan Gan
- College of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China.
| | - Xuemei Yang
- College of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China.
| | - Xinmei Lai
- College of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China.
| | - Yuxing Gao
- College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China.
| |
Collapse
|
45
|
Marwarha G, Schommer J, Lund J, Schommer T, Ghribi O. Palmitate-induced C/EBP homologous protein activation leads to NF-κB-mediated increase in BACE1 activity and amyloid beta genesis. J Neurochem 2018; 144:761-779. [PMID: 29315574 PMCID: PMC6371812 DOI: 10.1111/jnc.14292] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 11/29/2017] [Accepted: 12/23/2017] [Indexed: 12/12/2022]
Abstract
The etiology of Alzheimer's disease (AD) is egregiously comprehended, but epidemiological studies have posited that diets rich in the saturated fatty acid palmitic acid (palmitate) are a significant risk factor. The production and accumulation of amyloid beta peptide (Aβ) is considered the core pathological molecular event in the pathogenesis of AD. The rate-limiting step in Aβ genesis from amyloid-β precursor protein (AβPP) is catalyzed by the enzyme β-site amyloid precursor protein cleaving enzyme 1 (BACE1), the expression and enzymatic activity of which is significantly up-regulated in the AD brain. In this study, we determined the molecular mechanisms that potentially underlie the palmitate-induced up-regulation in BACE1 expression and augmented Aβ production. We demonstrate that a palmitate-enriched diet and exogenous palmitate treatment evoke an increase in BACE1 expression and activity leading to enhanced Aβ genesis in the mouse brain and SH-SY5Y-APPSwe cells, respectively, through the activation of the transcription factor NF-κB. Chromatin immunoprecipitation (ChIP) assays and luciferase reporter assays revealed that palmitate enhances BACE1 expression by increasing the binding of NF-κB in the BACE1 promoter followed by an enhancement in the transactivation of the BACE1 promoter. Elucidation and delineation of upstream molecular events unveiled a critical role of the endoplasmic reticulum stress-associated transcription factor, C/EBP homologous protein (CHOP) in the palmitate-induced NF-κB activation, as CHOP knock-down cells and Chop-/- mice do not exhibit the same degree of NF-κB activation in response to the palmitate challenge. Our study delineates a novel CHOP-NF-κB signaling pathway that mediates palmitate-induced up-regulation of BACE1 expression and Aβ genesis.
Collapse
Affiliation(s)
- Gurdeep Marwarha
- Department of Biomedical Sciences, School of Medicine & Health Sciences, University of North Dakota, Grand Forks, ND 58203
| | - Jared Schommer
- Department of Biomedical Sciences, School of Medicine & Health Sciences, University of North Dakota, Grand Forks, ND 58203
| | - Jonah Lund
- Department of Biomedical Sciences, School of Medicine & Health Sciences, University of North Dakota, Grand Forks, ND 58203
| | - Trevor Schommer
- Department of Biomedical Sciences, School of Medicine & Health Sciences, University of North Dakota, Grand Forks, ND 58203
| | - Othman Ghribi
- Department of Biomedical Sciences, School of Medicine & Health Sciences, University of North Dakota, Grand Forks, ND 58203
| |
Collapse
|
46
|
Fitz NF, Carter AY, Tapias V, Castranio EL, Kodali R, Lefterov I, Koldamova R. ABCA1 Deficiency Affects Basal Cognitive Deficits and Dendritic Density in Mice. J Alzheimers Dis 2018; 56:1075-1085. [PMID: 28106559 PMCID: PMC5302049 DOI: 10.3233/jad-161056] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
ATP-binding cassette transporter A1 (ABCA1) mediates cholesterol efflux to lipid-free apolipoproteins and regulates the generation of high density lipoproteins. Previously, we have shown that lack of Abca1 significantly increases amyloid deposition and cognitive deficits in Alzheimer’s disease model mice expressing human amyloid-β protein precursor (APP). The goal of this study was to determine if ABCA1 plays a role in memory deficits caused by amyloid-β (Aβ) oligomers and examine neurite architecture of pyramidal hippocampal neurons. Our results confirm previous findings that Abca1 deficiency significantly impairs spatial memory acquisition and retention in the Morris water maze and long-term memory in novel object recognition of APP transgenic mice at a stage of early amyloid pathology. Neither test demonstrated a significant difference between Abca1ko and wild-type (WT) mice. We also examined the effect of intra-hippocampal infused Aβ oligomers on cognitive performance of Abca1ko mice, compared to control infusion of scrambled Aβ peptide. Age-matched WT mice undergoing the same infusions were also used as controls. In this model system, we found a statistically significant difference between WT and Abca1ko mice infused with scrambled Aβ, suggesting that Abca1ko mice are vulnerable to the effect of mild stresses. Moreover, examination of neurite architecture in the hippocampi revealed a significant decrease in neurite length, number of neurite segments, and branches in Abca1ko mice when compared to WT mice. We conclude that mice lacking ABCA1 have basal cognitive deficits that prevent them from coping with additional stressors, which is in part due to impairment of neurite morphology in the hippocampus.
Collapse
Affiliation(s)
- Nicholas F Fitz
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Alexis Y Carter
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Victor Tapias
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Emilie L Castranio
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ravindra Kodali
- Department of Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Iliya Lefterov
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Radosveta Koldamova
- Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
47
|
Casali BT, Reed-Geaghan EG, Landreth GE. Nuclear receptor agonist-driven modification of inflammation and amyloid pathology enhances and sustains cognitive improvements in a mouse model of Alzheimer's disease. J Neuroinflammation 2018; 15:43. [PMID: 29448961 PMCID: PMC5815248 DOI: 10.1186/s12974-018-1091-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 02/07/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a highly prevalent neurodegenerative disorder characterized by pathological hallmarks of beta-amyloid plaque deposits, tau pathology, inflammation, and cognitive decline. Treatment remains a clinical obstacle due to lack of effective therapeutics. Agonists targeting nuclear receptors, such as bexarotene, reversed cognitive deficits regardless of treatment duration and age in murine models of AD. While bexarotene demonstrated marked efficacy in decreasing plaque levels following short-term treatment, prolonged treatment did not modulate plaque burden. This suggested that plaques might reform in mice treated chronically with bexarotene and that cessation of bexarotene treatment before plaques reform might alter amyloid pathology, inflammation, and cognition in AD mice. METHODS We utilized one-year-old APP/PS1 mice that were divided into two groups. We treated one group of mice for 2 weeks with bexarotene. The other group of mice was treated for 2 weeks with bexarotene followed by withdrawal of drug treatment for an additional 2 weeks. Cognition was evaluated using the novel-object recognition test either at the end of bexarotene treatment or the end of the withdrawal period. We then analyzed amyloid pathology and microgliosis at the conclusion of the study in both groups. RESULTS Bexarotene treatment enhanced cognition in APP/PS1 mice similar to previous findings. Strikingly, we observed sustained cognitive improvements in mice in which bexarotene treatment was discontinued for 2 weeks. We observed a sustained reduction in microgliosis and plaque burden following drug withdrawal exclusively in the hippocampus. CONCLUSIONS Our findings demonstrate that bexarotene selectively modifies aspects of neuroinflammation in a region-specific manner to reverse hippocampal-dependent cognitive deficits in AD mice and may provide insight to inform future studies with nuclear receptor agonists.
Collapse
Affiliation(s)
- Brad T. Casali
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106 USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Erin G. Reed-Geaghan
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106 USA
| | - Gary E. Landreth
- Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106 USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| |
Collapse
|
48
|
Salas IH, Weerasekera A, Ahmed T, Callaerts-Vegh Z, Himmelreich U, D'Hooge R, Balschun D, Saido TC, De Strooper B, Dotti CG. High fat diet treatment impairs hippocampal long-term potentiation without alterations of the core neuropathological features of Alzheimer disease. Neurobiol Dis 2018; 113:82-96. [PMID: 29427755 DOI: 10.1016/j.nbd.2018.02.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 02/04/2018] [Indexed: 01/25/2023] Open
Abstract
Type 2 diabetes (T2DM) and obesity might increase the risk for AD by 2-fold. Different attempts to model the effect of diet-induced diabetes on AD pathology in transgenic animal models, resulted in opposite conclusions. Here, we used a novel knock-in mouse model for AD, which, differently from other models, does not overexpress any proteins. Long-term high fat diet treatment triggers a reduction in hippocampal N-acetyl-aspartate/myo-inositol metabolites ratio and impairs long term potentiation in hippocampal acute slices. Interestingly, these alterations do not correlate with changes in the core neuropathological features of AD, i.e. amyloidosis and Tau hyperphosphorylation. The data suggest that AD phenotypes associated with high fat diet treatment seen in other models for AD might be exacerbated because of the overexpressing systems used to study the effects of familial AD mutations. Our work supports the increasing insight that knock-in mice might be more relevant models to study the link between metabolic disorders and AD.
Collapse
Affiliation(s)
- Isabel H Salas
- VIB Center for Brain and Disease Research, Leuven, Belgium; KU Leuven Department for Neurosciences, Leuven Institute for Neurodegenerative Disorders (LIND), KU Leuven, Leuven, Belgium
| | - Akila Weerasekera
- Biomedical MRI-Unit/MoSAIC, KU Leuven Campus Gasthuisberg, Leuven, Belgium
| | - Tariq Ahmed
- Laboratory of Biological Psychology, KU Leuven, Leuven, Belgium; Neurological Disorders Research Center, Doha, Qatar
| | | | - Uwe Himmelreich
- Biomedical MRI-Unit/MoSAIC, KU Leuven Campus Gasthuisberg, Leuven, Belgium
| | - Rudi D'Hooge
- Laboratory of Biological Psychology, KU Leuven, Leuven, Belgium
| | - Detlef Balschun
- Laboratory of Biological Psychology, KU Leuven, Leuven, Belgium
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Saitama, Japan
| | - Bart De Strooper
- VIB Center for Brain and Disease Research, Leuven, Belgium; KU Leuven Department for Neurosciences, Leuven Institute for Neurodegenerative Disorders (LIND), KU Leuven, Leuven, Belgium; UK Dementia Research Institute (DRI-UK), ION UCL, London, UK.
| | - Carlos G Dotti
- Centro de Biologıa Molecular 'Severo Ochoa' (CSIC/UAM), Madrid, Spain.
| |
Collapse
|
49
|
Zhang X, Lv C, An Y, Liu Q, Rong H, Tao L, Wang Y, Wang Y, Xiao R. Increased Levels of 27-Hydroxycholesterol Induced by Dietary Cholesterol in Brain Contribute to Learning and Memory Impairment in Rats. Mol Nutr Food Res 2018; 62. [PMID: 29193679 DOI: 10.1002/mnfr.201700531] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Revised: 11/08/2017] [Indexed: 12/22/2022]
Abstract
SCOPE Dietary cholesterol has been shown to play a role in the development of Alzheimer's disease (AD). It is proposed that oxysterol especially 27-hydroxycholesterol (27-OHC) may play a potential role in β-amyloid peptides (Aβ) production and accumulation during AD progression. METHODS AND RESULTS To investigate the mechanisms of dietary cholesterol and 27-OHC on learning and memory impairment, male Sprague-Dawley rats are fed with cholesterol diet with or without 27-OHC synthetase inhibitor (anastrozole) injection. The levels of cholesterol, 27-OHC, 24-hydroxycholesterol (24S-OHC), 7α-hydroxycholesterol, and 7β-hydroxycholesterol in plasma are determined; apolipoprotein A (ApoA), apolipoprotein B (ApoB), HDL-cholesterol (HDL-C), and LDL-cholesterol (LDL-C) in plasma or brain; CYP27A1 and CYP7A1 in liver and CYP46A1 and CYP7B1 in brain; cathepsin B, cathepsin D, and acid phosphatase in lysosome; and Aβ1-40 and Aβ1-42 in brain. Results show increased levels of 27-OHC (p < 0.01), LDL-C (p < 0.01), and ApoB (p < 0.01), and decreased level of HDL-C (p < 0.05) in plasma, upregulated CYP27A1 (p < 0.01) and CYP7A1 (p < 0.01) expression in liver, altered lysosomal function, and increased level of Aβ in brain (p < 0.05). CONCLUSIONS This study indicates that the mechanisms of dietary cholesterol on learning and memory impairment may be involved in cholesterol metabolism and lysosome function with the increase of plasma 27-OHC, thus resulting in Aβ formation and accumulation.
Collapse
Affiliation(s)
- Xiaona Zhang
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, China
| | - Chenyan Lv
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, China
| | - Yu An
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, China
| | - Quanri Liu
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, China
| | - Hongguo Rong
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, China
| | - Lingwei Tao
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, China
| | - Ying Wang
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, China
| | - Yushan Wang
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, China
| | - Rong Xiao
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, China
| |
Collapse
|
50
|
Kruse MS, Suarez LG, Coirini H. LXR activation increases the expression of GnRH AND αMSH in the rat hypothalamus in vivo. Neurosci Lett 2018; 664:20-27. [DOI: 10.1016/j.neulet.2017.11.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 10/11/2017] [Accepted: 11/07/2017] [Indexed: 11/25/2022]
|