1
|
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
|
2
|
Yazdi MK, Alavi MS, Roohbakhsh A. The role of ATP-binding cassette transporter G1 (ABCG1) in Alzheimer's disease: A review of the mechanisms. Basic Clin Pharmacol Toxicol 2024; 134:423-438. [PMID: 38275217 DOI: 10.1111/bcpt.13981] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 01/04/2024] [Accepted: 01/08/2024] [Indexed: 01/27/2024]
Abstract
The maintenance of cholesterol homeostasis is essential for central nervous system function. Consequently, factors that affect cholesterol homeostasis are linked to neurological disorders and pathologies. Among them, ATP-binding cassette transporter G1 (ABCG1) plays a significant role in atherosclerosis. However, its role in Alzheimer's disease (AD) is unclear. There is inconsistent information regarding ABCG1's role in AD. It can increase or decrease amyloid β (Aβ) levels in animals' brains. Clinical studies show that ABCG1 is involved in AD patients' impairment of cholesterol efflux capacity (CEC) in the cerebrospinal fluid (CSF). Lower Aβ levels in the CSF are correlated with ABCG1-mediated CEC dysfunction. ABCG1 modulates α-, β-, and γ-secretase activities in the plasma membrane and may affect Aβ production in the mitochondria-associated endoplasmic reticulum (ER) membrane (MAM) cell compartment. Despite contradictory findings regarding ABCG1's role in AD, this review shows that ABCG1 has a role in Aβ generation via modulation of membrane secretases. It is, however, necessary to investigate the underlying mechanism(s). ABCG1 may also contribute to AD pathology through its role in apoptosis and oxidative stress. As a result, ABCG1 plays a role in AD and is a candidate for drug development.
Collapse
Affiliation(s)
- Mohsen Karbasi Yazdi
- 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
- Department of Pharmacology, 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
|
3
|
Reyes-Reyes EM, Brown J, Trial MD, Chinnasamy D, Wiegand JP, Bradford D, Brinton RD, Rodgers KE. Vivaria housing conditions expose sex differences in brain oxidation, microglial activation, and immune system states in aged hAPOE4 mice. Exp Brain Res 2024; 242:543-557. [PMID: 38206365 PMCID: PMC10894770 DOI: 10.1007/s00221-023-06763-x] [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: 08/17/2023] [Accepted: 12/04/2023] [Indexed: 01/12/2024]
Abstract
Apolipoprotein E ε4 allele (APOE4) is the predominant genetic risk factor for late-onset Alzheimer's disease (AD). APOE4 mouse models have provided advances in the understanding of disease pathogenesis, but unaccounted variables like rodent housing status may hinder translational outcomes. Non-sterile aspects like food and bedding can be major sources of changes in rodent microflora. Alterations in intestinal microbial ecology can cause mucosal barrier impairment and increase pro-inflammatory signals. The present study examined the role of sterile and non-sterile food and housing on redox indicators and the immune status of humanized-APOE4 knock-in mice (hAPOe4). hAPOE4 mice were housed under sterile conditions until 22 months of age, followed by the transfer of a cohort of mice to non-sterile housing for 2 months. At 24 months of age, the redox/immunologic status was evaluated by flow cytometry/ELISA. hAPOE4 females housed under non-sterile conditions exhibited: (1) higher neuronal and microglial oxygen radical production and (2) lower CD68+ microglia (brain) and CD8+ T cells (periphery) compared to sterile-housed mice. In contrast, hAPOE4 males in non-sterile housing exhibited: (1) higher MHCII+ microglia and CD11b+CD4+ T cells (brain) and (2) higher CD11b+CD4+ T cells and levels of lipopolysaccharide-binding protein and inflammatory cytokines in the periphery relative to sterile-housed mice. This study demonstrated that sterile vs. non-sterile housing conditions are associated with the activation of redox and immune responses in the brain and periphery in a sex-dependent manner. Therefore, housing status may contribute to variable outcomes in both the brain and periphery.
Collapse
Affiliation(s)
- E M Reyes-Reyes
- Center for Innovation in Brain Science, University of Arizona, 1230 N. Cherry Ave, PO Box 210242, Tucson, AZ, 85721-0242, USA
| | - J Brown
- Center for Innovation in Brain Science, University of Arizona, 1230 N. Cherry Ave, PO Box 210242, Tucson, AZ, 85721-0242, USA
| | - M D Trial
- Center for Innovation in Brain Science, University of Arizona, 1230 N. Cherry Ave, PO Box 210242, Tucson, AZ, 85721-0242, USA
| | - D Chinnasamy
- Center for Innovation in Brain Science, University of Arizona, 1230 N. Cherry Ave, PO Box 210242, Tucson, AZ, 85721-0242, USA
| | - J P Wiegand
- Center for Innovation in Brain Science, University of Arizona, 1230 N. Cherry Ave, PO Box 210242, Tucson, AZ, 85721-0242, USA
| | - D Bradford
- Center for Innovation in Brain Science, University of Arizona, 1230 N. Cherry Ave, PO Box 210242, Tucson, AZ, 85721-0242, USA
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - R D Brinton
- Center for Innovation in Brain Science, University of Arizona, 1230 N. Cherry Ave, PO Box 210242, Tucson, AZ, 85721-0242, USA
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - K E Rodgers
- Center for Innovation in Brain Science, University of Arizona, 1230 N. Cherry Ave, PO Box 210242, Tucson, AZ, 85721-0242, USA.
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
4
|
Rahimpour A, Shahbazi B, Mafakher L. Discovery of small molecules from natural compound databases as potent retinoid X alpha receptor agonists to treat Alzheimer's disease. J Biomol Struct Dyn 2024:1-15. [PMID: 38373033 DOI: 10.1080/07391102.2024.2313166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 01/26/2024] [Indexed: 02/20/2024]
Abstract
Alzheimer's is characterized as a progressive neurodegenerative disease due to beta-amyloid accumulation in the brain. Some previous studies reported that RXR agonists could be effective in the treatment of Alzheimer's disease. There are currently numerous attempts being made to discover a natural RXR agonist that is more potent than 9-cis-retinoic acid (9CR). One of the most efficient resources for finding high-potential compounds is natural databases. In this study, 81215 compounds from the IB screen library as natural databases were docked against the RXR-alpha binding site. The best compounds discovered interact with the RXR-alpha binding site with a lower binding energy (-11 to -13 kcal/mol) than the binding energy of -10.94 kcal/mol for 9-cis, which means that these compounds could interact stronger with RXR-alpha than 9CR. All selected compounds could pass the blood-brain barrier. Physiochemical properties assessment indicated that all compounds passed Lipinski's rule and had the potential to be oral drug candidates. The stability of protein-ligand complexes during a timescale of 100 ns by Molecular Dynamics simulation demonstrated that all compounds could effectively interact with the RXR binding site. The molecular mechanics/Poisson-Boltzmann surface area (MM/PBSA) represented that all selected hit compounds had a better binding affinity to the alpha RXR binding site compared to 9CR, which means these hit compounds had potential drug candidates for the treatment of Alzheimer's disease. However, experimental assessment is needed to validate this result.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Alireza Rahimpour
- Islamic Azad University of Science and Research Branch Tehran, Tehran, Iran
| | - Behzad Shahbazi
- Molecular Medicine Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Ladan Mafakher
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
5
|
Takano-Kawabe K, Matoba K, Nakamura Y, Moriyama M. Low Density Lipoprotein Receptor-related Protein 2 Expression and Function in Cultured Astrocytes and Microglia. Neurochem Res 2024; 49:199-211. [PMID: 37702891 DOI: 10.1007/s11064-023-04022-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/23/2023] [Accepted: 08/28/2023] [Indexed: 09/14/2023]
Abstract
Activation of glial cells, astrocytes and microglia, has been observed in neurodegenerative diseases including Alzheimer's disease (AD). Amyloid β (Aβ), which is aggregated and the aggregation is detected as characteristic pathology in AD brain, is known to be produced by neurons and to activate glial cells. Clearance of Aβ from the brain via active transport system is important to prevent the accumulation and aggregation. Low density lipoprotein receptor-related protein 2 (LRP2/megalin) is an Aβ transporter. However, expression and contribution of LRP2 in astrocytes and microglia remain to be clarified. In the present study, we examined the expression of LRP2 and its roles in cultured astrocytes prepared from rat embryonic brain cortex and mouse microglial cell line BV-2. Both cultured rat astrocytes and BV-2 cells expressed LRP2 mRNA detected by RT-PCR. When lipopolysaccharide (LPS) or all-trans retinoic acid (ATRA) were added to BV-2 cells, LRP2 mRNA expression and uptake of microbeads, Aβ and insulin were increased. On the other hand, LPS decreased LRP2 expression and uptake of Aβ and insulin in cultured astrocytes. Knockdown of LRP2 using siRNA attenuated the LPS- or ATRA-increased uptake of microbeads, Aβ and insulin in BV-2 cells. These results suggest that LRP2 was expressed in both astrocytes and microglia and might be involved in endocytosis activities. Adequate control of LRP2 expression and function in astrocytes and microglia might regulate Aβ and insulin levels in brain and would be a potential target in AD pathology.
Collapse
Affiliation(s)
- Katsura Takano-Kawabe
- Laboratory of Integrative Physiology in Veterinary Sciences, Osaka Metropolitan University, 1-58, Rinku-Ourai Kita, Izumisano, Osaka, 598-8531, Japan.
| | - Kazuyuki Matoba
- Laboratory of Integrative Physiology in Veterinary Sciences, Osaka Metropolitan University, 1-58, Rinku-Ourai Kita, Izumisano, Osaka, 598-8531, Japan
| | - Yoichi Nakamura
- Laboratory of Integrative Physiology in Veterinary Sciences, Osaka Metropolitan University, 1-58, Rinku-Ourai Kita, Izumisano, Osaka, 598-8531, Japan
| | - Mitsuaki Moriyama
- Laboratory of Integrative Physiology in Veterinary Sciences, Osaka Metropolitan University, 1-58, Rinku-Ourai Kita, Izumisano, Osaka, 598-8531, Japan
| |
Collapse
|
6
|
Est CB, Murphy RM. An in vitro model for vitamin A transport across the human blood-brain barrier. eLife 2023; 12:RP87863. [PMID: 37934575 PMCID: PMC10629827 DOI: 10.7554/elife.87863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2023] Open
Abstract
Vitamin A, supplied by the diet, is critical for brain health, but little is known about its delivery across the blood-brain barrier (BBB). Brain microvascular endothelial-like cells (BMECs) differentiated from human-derived induced pluripotent stem cells (iPSCs) form a tight barrier that recapitulates many of the properties of the human BBB. We paired iPSC-derived BMECs with recombinant vitamin A serum transport proteins, retinol-binding protein (RBP), and transthyretin (TTR), to create an in vitro model for the study of vitamin A (retinol) delivery across the human BBB. iPSC-derived BMECs display a strong barrier phenotype, express key vitamin A metabolism markers, and can be used for quantitative modeling of retinol accumulation and permeation. Manipulation of retinol, RBP, and TTR concentrations, and the use of mutant RBP and TTR, yielded novel insights into the patterns of retinol accumulation in, and permeation across, the BBB. The results described herein provide a platform for deeper exploration of the regulatory mechanisms of retinol trafficking to the human brain.
Collapse
Affiliation(s)
- Chandler B Est
- Department of Chemical and Biological Engineering, University of WisconsinMadisonUnited States
| | - Regina M Murphy
- Department of Chemical and Biological Engineering, University of WisconsinMadisonUnited States
| |
Collapse
|
7
|
Liu Y, Wang P, Jin G, Shi P, Zhao Y, Guo J, Yin Y, Shao Q, Li P, Yang P. The novel function of bexarotene for neurological diseases. Ageing Res Rev 2023; 90:102021. [PMID: 37495118 DOI: 10.1016/j.arr.2023.102021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 07/13/2023] [Accepted: 07/20/2023] [Indexed: 07/28/2023]
Abstract
Bexarotene, a retinoid X receptor (RXR) agonist, is approved by FDA to treat cutaneous T-cell lymphoma. However, it has also demonstrated promising therapeutic potential for neurological diseases such as stroke, traumatic brain injury, Parkinson's disease, and particularly Alzheimer's disease(AD). In AD, bexarotene inhibits the production and aggregation of amyloid β (Aβ), activates Liver X Receptor/RXR heterodimers to increase lipidated apolipoprotein E to remove Aβ, mitigates the negative impact of Aβ, regulates neuroinflammation, and ultimately improves cognitive function. For other neurological diseases, its mechanisms of action include inhibiting inflammatory responses, up-regulating microglial phagocytosis, and reducing misfolded protein aggregation, all of which aid in alleviating neurological damage. Here, we briefly discuss the characteristics, applications, and adverse effects of bexarotene, summarize its pharmacological mechanisms and therapeutic results in various neurological diseases, and elaborate on the problems encountered in preclinical research, with the aim of providing help for the further application of bexarotene in central nervous system diseases.
Collapse
Affiliation(s)
- Yangtao Liu
- College of Pharamacy, Xinxiang Medical University, Xinxiang, China; Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China; College of Third Clinical, Xinxiang Medical University, Xinxiang, China
| | - Pengwei Wang
- Department of Pharmacy, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang Medical University, Weihui, China
| | - Guofang Jin
- College of Pharamacy, Xinxiang Medical University, Xinxiang, China; Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China
| | - Peijie Shi
- College of Pharamacy, Xinxiang Medical University, Xinxiang, China; Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China; Xinxiang First People's Hospital, The Affiliated People's Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yonghui Zhao
- Xinxiang First People's Hospital, The Affiliated People's Hospital of Xinxiang Medical University, Xinxiang, China
| | - Jiayi Guo
- College of Pharamacy, Xinxiang Medical University, Xinxiang, China; Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China
| | - Yaling Yin
- School of Basic Medical Sciences, Department of Physiology and Pathophysiology, Sino-UK Joint Laboratory of Brain Function and Injury and Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, China
| | - Qianhang Shao
- Department of Pharmacy, People's Hospital of Peking University, Beijing, China.
| | - Peng Li
- College of Pharamacy, Xinxiang Medical University, Xinxiang, China; Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.
| | - Pengfei Yang
- College of Pharamacy, Xinxiang Medical University, Xinxiang, China; Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.
| |
Collapse
|
8
|
Bi G, Liang J, Shan G, Bian Y, Chen Z, Huang Y, Lu T, Li M, Besskaya V, Zhao M, Fan H, Wang Q, Gan B, Zhan C. Retinol Saturase Mediates Retinoid Metabolism to Impair a Ferroptosis Defense System in Cancer Cells. Cancer Res 2023; 83:2387-2404. [PMID: 37184371 DOI: 10.1158/0008-5472.can-22-3977] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 03/22/2023] [Accepted: 05/10/2023] [Indexed: 05/16/2023]
Abstract
Ferroptosis is an iron-dependent form of regulated cell death induced by the lethal overload of lipid peroxides in cellular membranes. In recent years, modulating ferroptosis has gained attention as a potential therapeutic approach for tumor suppression. In the current study, retinol saturase (RETSAT) was identified as a significant ferroptosis mediator using a publicly accessible CRISPR/Cas9 screening dataset. RETSAT depletion protected tumor cells from lipid peroxidation and subsequent cell death triggered by various ferroptosis inducers. Furthermore, exogenous supplementation with retinoids, including retinol (the substrate of RETSAT) and its derivatives retinal and retinoic acid, also suppressed ferroptosis, whereas the product of RETSAT, 13, 14-dihydroretinol, failed to do so. As effective radical-trapping antioxidant, retinoids protected the lipid membrane from autoxidation and subsequent fragmentation, thus terminating the cascade of ferroptosis. Pseudotargeted lipidomic analysis identified an association between retinoid regulation of ferroptosis and lipid metabolism. Retinoic acid, but not 13, 14-dihydroretinoic acid, interacted with its nuclear receptor and activated transcription of stearoyl-CoA desaturase, which introduces the first double bond into saturated fatty acid and thus catalyzes the generation of monounsaturated fatty acid, a known ferroptosis suppressor. Therefore, RETSAT promotes ferroptosis by transforming retinol to 13, 14-dihydroretinol, thereby turning a strong anti-ferroptosis regulator into a relatively weak one. SIGNIFICANCE Retinoids have ferroptosis-protective properties and can be metabolized by RETSAT to promote ferroptosis, suggesting the possibility of targeting retinoid metabolism in cancer as a treatment strategy to trigger ferroptosis.
Collapse
Affiliation(s)
- Guoshu Bi
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Jiaqi Liang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Guangyao Shan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Yunyi Bian
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Zhencong Chen
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Yiwei Huang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Tao Lu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Ming Li
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Valeria Besskaya
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Mengnan Zhao
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Hong Fan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Qun Wang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Boyi Gan
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Cheng Zhan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| |
Collapse
|
9
|
Jakaria M, Belaidi AA, Bush AI, Ayton S. Vitamin A metabolites inhibit ferroptosis. Biomed Pharmacother 2023; 164:114930. [PMID: 37236031 DOI: 10.1016/j.biopha.2023.114930] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/18/2023] [Accepted: 05/22/2023] [Indexed: 05/28/2023] Open
Abstract
Vitamin A (retinol) is a lipid-soluble vitamin that acts as a precursor for several bioactive compounds, such as retinaldehyde (retinal) and isomers of retinoic acid. Retinol and all-trans-retinoic acid (atRA) penetrate the blood-brain barrier and are reported to be neuroprotective in several animal models. We characterised the impact of retinol and its metabolites, all-trans-retinal (atRAL) and atRA, on ferroptosis-a programmed cell death caused by iron-dependent phospholipid peroxidation. Ferroptosis was induced by erastin, buthionine sulfoximine or RSL3 in neuronal and non-neuronal cell lines. We found that retinol, atRAL and atRA inhibited ferroptosis with a potency superior to α-tocopherol, the canonical anti-ferroptotic vitamin. In contrast, we found that antagonism of endogenous retinol with anhydroretinol sensitises ferroptosis induced in neuronal and non-neuronal cell lines. Retinol and its metabolites atRAL and atRA directly interdict lipid radicals in ferroptosis since these compounds displayed radical trapping properties in a cell-free assay. Vitamin A, therefore, complements other anti-ferroptotic vitamins, E and K; metabolites of vitamin A, or agents that alter their levels, may be potential therapeutics for diseases where ferroptosis is implicated.
Collapse
Affiliation(s)
- Md Jakaria
- The Florey Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Abdel A Belaidi
- The Florey Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Ashley I Bush
- The Florey Institute, The University of Melbourne, Parkville, Victoria, Australia.
| | - Scott Ayton
- The Florey Institute, The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
10
|
Est CB, Murphy RM. An in vitro model for vitamin A transport across the human blood-brain barrier. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.11.536348. [PMID: 37090623 PMCID: PMC10120720 DOI: 10.1101/2023.04.11.536348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Vitamin A, supplied by the diet, is critical for brain health, but little is known about its delivery across the blood-brain barrier (BBB). Brain microvascular endothelial-like cells (BMECs) differentiated from human-derived induced pluripotent stem cells (iPSC) form a tight barrier that recapitulates many of the properties of the human BBB. We paired iPSC-derived BMECs with recombinant vitamin A serum transport proteins, retinol binding protein (RBP) and transthyretin (TTR), to create an in vitro model for the study of vitamin A (retinol) delivery across the human BBB. iPSC-derived BMECs display a strong barrier phenotype, express key vitamin A metabolism markers and can be used for quantitative modeling of retinol accumulation and permeation. Manipulation of retinol, RBP and TTR concentrations, and the use of mutant RBP and TTR, yielded novel insights into the patterns of retinol accumulation in, and permeation across, the BBB. The results described herein provide a platform for deeper exploration of the regulatory mechanisms of retinol trafficking to the human brain.
Collapse
Affiliation(s)
| | - Regina M. Murphy
- Department of Chemical and Biological Engineering, University of Wisconsin – Madison 1415 Engineering Dr., Madison, WI 53706
| |
Collapse
|
11
|
Kessler K, Giannisis A, Bial G, Foquet L, Nielsen HM, Raber J. Behavioral and cognitive performance of humanized APOEε3/ε3 liver mice in relation to plasma apolipoprotein E levels. Sci Rep 2023; 13:1728. [PMID: 36720957 PMCID: PMC9889814 DOI: 10.1038/s41598-023-28165-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 01/13/2023] [Indexed: 02/01/2023] Open
Abstract
Plasma apolipoprotein E levels were previously associated with the risk of developing Alzheimer's disease (AD), levels of cerebrospinal fluid AD biomarkers, cognition and imaging brain measures. Outside the brain, the liver is the primary source of apoE and liver transplantation studies have demonstrated that liver-derived apoE does not cross the blood-brain-barrier. How hepatic apoE may be implicated in behavioral and cognitive performance is not clear. In the current study, we behaviorally tested FRGN mice with humanized liver harboring the ε3/ε3 genotype (E3-human liver (HL)) and compared their behavioral and cognitive performance with that of age-matched ε3/ε3 targeted replacement (E3-TR) mice, the latter produces human apoE3 throughout the body whereas the E3-HL mice endogenously produce human apoE3 only in the liver. We also compared the liver weights and plasma apoE levels, and assessed whether plasma apoE levels were correlated with behavioral or cognitive measures in both models. E3-HL were more active but performed cognitively worse than E3-TR mice. E3-HL mice moved more in the open field containing objects, showed higher activity levels in the Y maze, showed higher activity levels during the baseline period in the fear conditioning test than E3-TR mice, and swam faster than E3-TR mice during training to locate the visible platform in the water maze. However, E3-HL mice showed reduced spatial memory retention in the water maze and reduced fear learning and contextual and cued fear memory than E3-TR mice. Liver weights were greater in E3-HL than E3-TR mice and sex-dependent only in the latter model. Plasma apoE3 levels were similar to those found in humans and comparable in female and male E3-TR mice but higher in female E3-HL mice. Finally, we found correlations between plasma apoE levels and behavioral and cognitive measures which were predominantly model-dependent. Our study demonstrates mouse-model dependent associations between plasma apoE levels, behavior and cognition in an 'AD-neutral' setting and suggests that a humanized liver might be sufficient to induce mouse behavioral and cognitive phenotypes.
Collapse
Affiliation(s)
- Kat Kessler
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Andreas Giannisis
- Department of Biochemistry and Biophysics, Stockholm University, 10691, Stockholm, Sweden
| | - Greg Bial
- Yecuris Corporation, Tualatin, OR, 97062, USA
| | | | - Henrietta M Nielsen
- Department of Biochemistry and Biophysics, Stockholm University, 10691, Stockholm, Sweden.
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239, USA. .,Departments of Neurology and Radiation Medicine, Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, 97239, USA.
| |
Collapse
|
12
|
Mathew A, Balaji E V, Pai SRK, Kishore A, Pai V, Pemmireddy R, K S C. Current Drug Targets in Alzheimer's Associated Memory Impairment: A Comprehensive Review. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2023; 22:255-275. [PMID: 35366787 DOI: 10.2174/1871527321666220401124719] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 12/17/2021] [Accepted: 01/19/2022] [Indexed: 12/16/2022]
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia among geriatrics. It is a progressive, degenerative neurologic disorder that causes memory and cognition loss. The accumulation of amyloid fibrils and neurofibrillary tangles in the brain of AD patients is a distinguishing feature of the disease. Therefore, most of the current therapeutic goals are targeting inhibition of beta-amyloid synthesis and aggregation as well as tau phosphorylation and aggregation. There is also a loss of the cholinergic neurons in the basal forebrain, and first-generation therapeutic agents were primarily focused on compensating for this loss of neurons. However, cholinesterase inhibitors can only alleviate cognitive symptoms of AD and cannot reduce the progression of the disease. Understanding the molecular and cellular changes associated with AD pathology has advanced significantly in recent decades. The etiology of AD is complex, with a substantial portion of sporadic AD emerging from unknown reasons and a lesser proportion of early-onset familial AD (FAD) caused by a mutation in several genes, such as the amyloid precursor protein (APP), presenilin 1 (PS1), and presenilin 2 (PS2) genes. Hence, efforts are being made to discover novel strategies for these targets for AD therapy. A new generation of AChE and BChE inhibitors is currently being explored and evaluated in human clinical trials for AD symptomatic treatment. Other approaches for slowing the progression of AD include serotonergic modulation, H3 receptor antagonism, phosphodiesterase, COX-2, and MAO-B inhibition. The present review provides an insight into the possible therapeutic strategies and their molecular mechanisms, enlightening the perception of classical and future treatment approaches.
Collapse
Affiliation(s)
- Anna Mathew
- Department of Pharmacognosy, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka-576104, India
| | - Vignesh Balaji E
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka-576104, India
| | - Sreedhara Ranganath K Pai
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka-576104, India
| | - Anoop Kishore
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka-576104, India
| | - Vasudev Pai
- Department of Pharmacognosy, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka-576104, India
| | - Ramadevi Pemmireddy
- Department of Pharmacognosy, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka-576104, India
| | - Chandrashekar K S
- Department of Pharmacognosy, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka-576104, India
| |
Collapse
|
13
|
Huang X, Wang YJ, Xiang Y. Bidirectional communication between brain and visceral white adipose tissue: Its potential impact on Alzheimer's disease. EBioMedicine 2022; 84:104263. [PMID: 36122553 PMCID: PMC9490488 DOI: 10.1016/j.ebiom.2022.104263] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 08/21/2022] [Accepted: 08/24/2022] [Indexed: 11/20/2022] Open
Abstract
A variety of axes between brain and abdominal organs have been reported, but the interaction between brain and visceral white adipose tissue (vWAT) remains unclear. In this review, we summarized human studies on the association between brain and vWAT, and generalized their interaction and the underlying mechanisms according to animal and cell experiments. On that basis, we come up with the concept of the brain-vWAT axis (BVA). Furthermore, we analyzed the potential mechanisms of involvement of BVA in the pathogenesis of Alzheimer's disease (AD), including vWAT-derived fatty acids, immunological properties of vWAT, vWAT-derived retinoic acid and vWAT-regulated insulin resistance. The proposal of BVA may expand our understanding to some extent of how the vWAT impacts on brain health and diseases, and provide a novel approach to study the pathogenesis and treatment strategies of neurodegenerative disorders.
Collapse
|
14
|
Giannisis A, Patra K, Edlund AK, Nieto LA, Benedicto-Gras J, Moussaud S, de la Rosa A, Twohig D, Bengtsson T, Fu Y, Bu G, Bial G, Foquet L, Hammarstedt C, Strom S, Kannisto K, Raber J, Ellis E, Nielsen HM. Brain integrity is altered by hepatic APOE ε4 in humanized-liver mice. Mol Psychiatry 2022; 27:3533-3543. [PMID: 35418601 PMCID: PMC9708568 DOI: 10.1038/s41380-022-01548-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/21/2022] [Accepted: 03/23/2022] [Indexed: 12/12/2022]
Abstract
Liver-generated plasma apolipoprotein E (apoE) does not enter the brain but nonetheless correlates with Alzheimer's disease (AD) risk and AD biomarker levels. Carriers of APOEε4, the strongest genetic AD risk factor, exhibit lower plasma apoE and altered brain integrity already at mid-life versus non-APOEε4 carriers. Whether altered plasma liver-derived apoE or specifically an APOEε4 liver phenotype promotes neurodegeneration is unknown. Here we investigated the brains of Fah-/-, Rag2-/-, Il2rg-/- mice on the Non-Obese Diabetic (NOD) background (FRGN) with humanized-livers of an AD risk-associated APOE ε4/ε4 versus an APOE ε2/ε3 genotype. Reduced endogenous mouse apoE levels in the brains of APOE ε4/ε4 liver mice were accompanied by various changes in markers of synaptic integrity, neuroinflammation and insulin signaling. Plasma apoE4 levels were associated with unfavorable changes in several of the assessed markers. These results propose a previously unexplored role of the liver in the APOEε4-associated risk of neurodegenerative disease.
Collapse
Affiliation(s)
- Andreas Giannisis
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, 10691, Sweden
| | - Kalicharan Patra
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, 10691, Sweden
| | - Anna K Edlund
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, 10691, Sweden
| | - Lur Agirrezabala Nieto
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, 10691, Sweden
| | - Joan Benedicto-Gras
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, 10691, Sweden
| | - Simon Moussaud
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, 10691, Sweden
| | - Andrés de la Rosa
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, 10691, Sweden
| | - Daniel Twohig
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, 10691, Sweden
| | - Tore Bengtsson
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm, 10691, Sweden
| | - Yuan Fu
- Department of Neuroscience, Mayo Clinic College of Medicine, Jacksonville, FL, 32224, USA
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic College of Medicine, Jacksonville, FL, 32224, USA
| | - Greg Bial
- Yecuris Corporation, Tualatin, OR, 97062, USA
| | | | - Christina Hammarstedt
- Department of Laboratory Medicine (LABMED), Karolinska Institutet, Stockholm, 17177, Sweden
| | - Stephen Strom
- Department of Laboratory Medicine (LABMED), Karolinska Institutet, Stockholm, 17177, Sweden
| | - Kristina Kannisto
- Department of Laboratory Medicine (LABMED), Karolinska Institutet, Stockholm, 17177, Sweden
| | - Jacob Raber
- Departments of Behavioral Neuroscience, Neurology, and Radiation Medicine, and Division of Neuroscience, ONPPRC, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Ewa Ellis
- Department of Clinical Science, Intervention and Technology, (CLINTEC), Division of Transplantation surgery, Karolinska Institutet, Huddinge, 14152, Sweden
| | - Henrietta M Nielsen
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, 10691, Sweden.
| |
Collapse
|
15
|
Liu CC, Zhao J, Fu Y, Inoue Y, Ren Y, Chen Y, Doss SV, Shue F, Jeevaratnam S, Bastea L, Wang N, Martens YA, Qiao W, Wang M, Zhao N, Jia L, Yamazaki Y, Yamazaki A, Rosenberg CL, Wang Z, Kong D, Li Z, Kuchenbecker LA, Trottier ZA, Felton L, Rogers J, Quicksall ZS, Linares C, Knight J, Chen Y, Kurti A, Kanekiyo T, Fryer JD, Asmann YW, Storz P, Wang X, Peng J, Zhang B, Kim BYS, Bu G. Peripheral apoE4 enhances Alzheimer's pathology and impairs cognition by compromising cerebrovascular function. Nat Neurosci 2022; 25:1020-1033. [PMID: 35915180 PMCID: PMC10009873 DOI: 10.1038/s41593-022-01127-0] [Citation(s) in RCA: 78] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 06/22/2022] [Indexed: 12/21/2022]
Abstract
The ε4 allele of the apolipoprotein E (APOE) gene, a genetic risk factor for Alzheimer's disease, is abundantly expressed in both the brain and periphery. Here, we present evidence that peripheral apoE isoforms, separated from those in the brain by the blood-brain barrier, differentially impact Alzheimer's disease pathogenesis and cognition. To evaluate the function of peripheral apoE, we developed conditional mouse models expressing human APOE3 or APOE4 in the liver with no detectable apoE in the brain. Liver-expressed apoE4 compromised synaptic plasticity and cognition by impairing cerebrovascular functions. Plasma proteome profiling revealed apoE isoform-dependent functional pathways highlighting cell adhesion, lipoprotein metabolism and complement activation. ApoE3 plasma from young mice improved cognition and reduced vessel-associated gliosis when transfused into aged mice, whereas apoE4 compromised the beneficial effects of young plasma. A human induced pluripotent stem cell-derived endothelial cell model recapitulated the plasma apoE isoform-specific effect on endothelial integrity, further supporting a vascular-related mechanism. Upon breeding with amyloid model mice, liver-expressed apoE4 exacerbated brain amyloid pathology, whereas apoE3 reduced it. Our findings demonstrate pathogenic effects of peripheral apoE4, providing a strong rationale for targeting peripheral apoE to treat Alzheimer's disease.
Collapse
Affiliation(s)
- Chia-Chen Liu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA.
| | - Jing Zhao
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Yuan Fu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Yasuteru Inoue
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Yingxue Ren
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL, USA
| | - Yuanxin Chen
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Sydney V Doss
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Francis Shue
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | - Ligia Bastea
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Na Wang
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Yuka A Martens
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Wenhui Qiao
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Minghui Wang
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Na Zhao
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Lin Jia
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Yu Yamazaki
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Akari Yamazaki
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | - Zhen Wang
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Dehui Kong
- Department of Biology, University of North Dakota, Grand Forks, ND, USA
| | - Zonghua Li
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | | | - Lindsey Felton
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Justin Rogers
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | - Cynthia Linares
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Joshua Knight
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Yixing Chen
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Aishe Kurti
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | - John D Fryer
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Yan W Asmann
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL, USA
| | - Peter Storz
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Xusheng Wang
- Department of Biology, University of North Dakota, Grand Forks, ND, USA
| | - Junmin Peng
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Betty Y S Kim
- Department of Neurosurgery, The Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA.
| |
Collapse
|
16
|
Abstract
The brain, as one of the most lipid-rich organs, heavily relies on lipid transport and distribution to maintain homeostasis and neuronal function. Lipid transport mediated by lipoprotein particles, which are complex structures composed of apolipoproteins and lipids, has been thoroughly characterized in the periphery. Although lipoproteins in the central nervous system (CNS) were reported over half a century ago, the identification of APOE4 as the strongest genetic risk factor for Alzheimer's disease has accelerated investigation of the biology and pathobiology of lipoproteins in the CNS. This review provides an overview of the different components of lipoprotein particles, in particular apolipoproteins, and their involvements in both physiological functions and pathological mechanisms in the CNS.
Collapse
Affiliation(s)
| | - Yuka A Martens
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA;
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA;
| |
Collapse
|
17
|
Jin Y, Li F, Sonoustoun B, Kondru NC, Martens YA, Qiao W, Heckman MG, Ikezu TC, Li Z, Burgess JD, Amerna D, O’Leary J, DeTure MA, Zhao J, McLean PJ, Dickson DW, Ross OA, Bu G, Zhao N. APOE4 exacerbates α-synuclein seeding activity and contributes to neurotoxicity in Alzheimer's disease with Lewy body pathology. Acta Neuropathol 2022; 143:641-662. [PMID: 35471463 PMCID: PMC9107450 DOI: 10.1007/s00401-022-02421-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 04/15/2022] [Accepted: 04/16/2022] [Indexed: 01/17/2023]
Abstract
Approximately half of Alzheimer's disease (AD) brains have concomitant Lewy pathology at autopsy, suggesting that α-synuclein (α-SYN) aggregation is a regulated event in the pathogenesis of AD. Genome-wide association studies revealed that the ε4 allele of the apolipoprotein E (APOE4) gene, the strongest genetic risk factor for AD, is also the most replicated genetic risk factor for Lewy body dementia (LBD), signifying an important role of APOE4 in both amyloid-β (Aβ) and α-SYN pathogenesis. How APOE4 modulates α-SYN aggregation in AD is unclear. In this study, we aimed to determine how α-SYN is associated with AD-related pathology and how APOE4 impacts α-SYN seeding and toxicity. We measured α-SYN levels and their association with other established AD-related markers in brain samples from autopsy-confirmed AD patients (N = 469), where 54% had concomitant LB pathology (AD + LB). We found significant correlations between the levels of α-SYN and those of Aβ40, Aβ42, tau and APOE, particularly in insoluble fractions of AD + LB. Using a real-time quaking-induced conversion (RT-QuIC) assay, we measured the seeding activity of soluble α-SYN and found that α-SYN seeding was exacerbated by APOE4 in the AD cohort, as well as a small cohort of autopsy-confirmed LBD brains with minimal Alzheimer type pathology. We further fractionated the soluble AD brain lysates by size exclusion chromatography (SEC) ran on fast protein liquid chromatography (FPLC) and identified the α-SYN species (~ 96 kDa) that showed the strongest seeding activity. Finally, using human induced pluripotent stem cell (iPSC)-derived neurons, we showed that amplified α-SYN aggregates from AD + LB brain of patients with APOE4 were highly toxic to neurons, whereas the same amount of α-SYN monomer was not toxic. Our findings suggest that the presence of LB pathology correlates with AD-related pathologies and that APOE4 exacerbates α-SYN seeding activity and neurotoxicity, providing mechanistic insight into how APOE4 affects α-SYN pathogenesis in AD.
Collapse
|
18
|
Martens YA, Zhao N, Liu CC, Kanekiyo T, Yang AJ, Goate AM, Holtzman DM, Bu G. ApoE Cascade Hypothesis in the pathogenesis of Alzheimer's disease and related dementias. Neuron 2022; 110:1304-1317. [PMID: 35298921 PMCID: PMC9035117 DOI: 10.1016/j.neuron.2022.03.004] [Citation(s) in RCA: 123] [Impact Index Per Article: 61.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 02/08/2022] [Accepted: 03/01/2022] [Indexed: 12/21/2022]
Abstract
The ε4 allele of the apolipoprotein E gene (APOE4) is a strong genetic risk factor for Alzheimer's disease (AD) and several other neurodegenerative conditions, including Lewy body dementia (LBD). The three APOE alleles encode protein isoforms that differ from one another only at amino acid positions 112 and 158: apoE2 (C112, C158), apoE3 (C112, R158), and apoE4 (R112, R158). Despite progress, it remains unclear how these small amino acid differences in apoE sequence among the three isoforms lead to profound effects on aging and disease-related pathways. Here, we propose a novel "ApoE Cascade Hypothesis" in AD and age-related cognitive decline, which states that the biochemical and biophysical properties of apoE impact a cascade of events at the cellular and systems levels, ultimately impacting aging-related pathogenic conditions including AD. As such, apoE-targeted therapeutic interventions are predicted to be more effective by addressing the biochemical phase of the cascade.
Collapse
Affiliation(s)
- Yuka A Martens
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Na Zhao
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Chia-Chen Liu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | - Austin J Yang
- Division of Neuroscience, National Institute on Aging, Bethesda, MD, USA
| | - Alison M Goate
- Ronald M. Loeb Center for Alzheimer's Disease, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA.
| |
Collapse
|
19
|
Pouso MR, Cairrao E. Effect of retinoic acid on the neurovascular unit: A review. Brain Res Bull 2022; 184:34-45. [DOI: 10.1016/j.brainresbull.2022.03.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 03/08/2022] [Accepted: 03/21/2022] [Indexed: 12/12/2022]
|
20
|
Sanjay, Kim JY. Anti-inflammatory effects of 9-cis-retinoic acid on β-amyloid treated human microglial cells. EUR J INFLAMM 2022. [DOI: 10.1177/1721727x221143651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Objective: Alzheimer’s disease (AD) is a neurodegenerative disease that abolishes cognitive and analytical abilities to perform basic day-to-day tasks. Microglia are involved in AD-associated neuroinflammation in response to amyloid β‐peptide (Aβ). This study focused on observing the immunomodulatory effects of 9-cis-retinoic acid (9-Cis-RA) the active metabolite of vitamin A, on Aβ-treated human microglial HMO6 cells. Methods: HMO6 cells were treated with Aβ42 in the absence or presence of 9-cis-RA, and the expression of M1-and M2-associated molecules, Toll like receptors (TLRs), and triggering receptor expressed on myeloid cells 2 (TREM2) were examined. Results: The levels of M1-markers [cluster of differentiation (CD86) and inducible nitric oxide synthase (iNOS)] and -cytokines [tumor necrosis factor (TNF-α), interleukin (IL)-6, and IL-1β], inflammatory receptors (TLR2 and TLR4), and reactive oxygen species increased significantly in Aβ-treated HMO6 cells. In contrast, the levels of M2-markers (CD206 and arginase-1) and -cytokines (IL-10, IL-4, and C-C motif chemokine ligand 17) the anti-inflammatory receptor TLR10 was significantly suppressed. However, 9-cis-RA treatment reversed the Aβ-induced upregulation of expression of M1-associated molecules and upregulated the expression of M2-associated molecules. Moreover, 9-cis-RA treatment augmented Aβ uptake by HMO6 cells, possibly by increasing the cell surface protein levels of TREM2, which is a receptor of Aβ that promotes Aβ phagocytosis by microglia. Conclusion: Our results suggest that 9-cis-RA is a potential therapeutic agent for AD.
Collapse
Affiliation(s)
- Sanjay
- Department of Life Science, Gachon University, Seongnam, Korea
| | - Jae Young Kim
- Department of Life Science, Gachon University, Seongnam, Korea
| |
Collapse
|
21
|
Thiel W, Esposito EJ, Findley AP, Blume ZI, Mitchell DM. Modulation of retinoid-X-receptors differentially regulates expression of apolipoprotein genes apoc1 and apoeb by zebrafish microglia. Biol Open 2021; 11:273656. [PMID: 34878094 PMCID: PMC8822359 DOI: 10.1242/bio.058990] [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: 08/13/2021] [Accepted: 11/30/2021] [Indexed: 11/20/2022] Open
Abstract
Transcriptome analyses performed in both human and zebrafish indicate strong expression of Apoe and Apoc1 by microglia. Apoe expression by microglia is well appreciated, but Apoc1 expression has not been well-examined. PPAR/RXR and LXR/RXR receptors appear to regulate expression of the apolipoprotein gene cluster in macrophages, but a similar role in microglia in vivo has not been studied. Here, we characterized microglial expression of apoc1 in the zebrafish central nervous system (CNS) in situ and demonstrate that in the CNS, apoc1 expression is unique to microglia. We then examined the effects of PPAR/RXR and LXR/RXR modulation on microglial expression of apoc1 and apoeb during early CNS development using a pharmacological approach. Changes in apoc1 and apoeb transcripts in response to pharmacological modulation were quantified by RT-qPCR in whole heads, and in individual microglia using hybridization chain reaction (HCR) in situ hybridization. We found that expression of apoc1 and apoeb by microglia were differentially regulated by LXR/RXR and PPAR/RXR modulating compounds, respectively, during development. Our results also suggest RXR receptors could be involved in endogenous induction of apoc1 expression by microglia. Collectively, our work supports the use of zebrafish to better understand regulation and function of these apolipoproteins in the CNS. Summary: Here we investigate expression of two apolipoprotein genes by microglia in the zebrafish model during normal development, and in contexts of pharmacological manipulations that target candidate regulatory receptors.
Collapse
Affiliation(s)
- Whitney Thiel
- Biological Sciences, University of Idaho, Moscow, ID 83844, Russia
| | - Emma J Esposito
- Biological Sciences, University of Idaho, Moscow, ID 83844, Russia
| | - Anna P Findley
- Biological Sciences, University of Idaho, Moscow, ID 83844, Russia
| | - Zachary I Blume
- Biological Sciences, University of Idaho, Moscow, ID 83844, Russia
| | - Diana M Mitchell
- Biological Sciences, University of Idaho, Moscow, ID 83844, Russia
| |
Collapse
|
22
|
Liu CC, Murray ME, Li X, Zhao N, Wang N, Heckman MG, Shue F, Martens Y, Li Y, Raulin AC, Rosenberg CL, Doss SV, Zhao J, Wren MC, Jia L, Ren Y, Ikezu TC, Lu W, Fu Y, Caulfield T, Trottier ZA, Knight J, Chen Y, Linares C, Wang X, Kurti A, Asmann YW, Wszolek ZK, Smith GE, Vemuri P, Kantarci K, Knopman DS, Lowe VJ, Jack CR, Parisi JE, Ferman TJ, Boeve BF, Graff-Radford NR, Petersen RC, Younkin SG, Fryer JD, Wang H, Han X, Frieden C, Dickson DW, Ross OA, Bu G. APOE3-Jacksonville (V236E) variant reduces self-aggregation and risk of dementia. Sci Transl Med 2021; 13:eabc9375. [PMID: 34586832 PMCID: PMC8824726 DOI: 10.1126/scitranslmed.abc9375] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Apolipoprotein E (APOE) genetic variants have been shown to modify Alzheimer’s disease (AD) risk. We previously identified an APOE3 variant (APOE3-V236E), named APOE3-Jacksonville (APOE3-Jac), associated with healthy brain aging and reduced risk for AD and dementia with Lewy bodies (DLB). Herein, we resolved the functional mechanism by which APOE3-Jac reduces APOE aggregation and enhances its lipidation in human brains, as well as in cellular and biochemical assays. Compared to APOE3, expression of APOE3-Jac in astrocytes increases several classes of lipids in the brain including phosphatidylserine, phosphatidylethanolamine, phosphatidic acid, and sulfatide, critical for synaptic functions. Mice expressing APOE3-Jac have reduced amyloid pathology, plaque-associated immune responses, and neuritic dystrophy. The V236E substitution is also sufficient to reduce the aggregation of APOE4, whose gene allele is a major genetic risk factor for AD and DLB. These findings suggest that targeting APOE aggregation might be an effective strategy for treating a subgroup of individuals with AD and DLB.
Collapse
Affiliation(s)
- Chia-Chen Liu
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | | | - Xia Li
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Na Zhao
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Na Wang
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Michael G. Heckman
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Jacksonville, Florida, USA
| | - Francis Shue
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Yuka Martens
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Yonghe Li
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | | | | | - Sydney V. Doss
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Jing Zhao
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Melissa C. Wren
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Lin Jia
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Yingxue Ren
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Jacksonville, Florida, USA
| | | | - Wenyan Lu
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Yuan Fu
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Thomas Caulfield
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | | | - Joshua Knight
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Yixing Chen
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Cynthia Linares
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Xue Wang
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Jacksonville, Florida, USA
| | - Aishe Kurti
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Yan W. Asmann
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Jacksonville, Florida, USA
| | | | - Glenn E. Smith
- Department of Psychiatry and Psychology Mayo Clinic, Rochester, Minnesota, USA
| | | | - Kejal Kantarci
- Department of Radiology Mayo Clinic, Rochester, Minnesota, USA
| | | | - Val J. Lowe
- Department of Radiology Mayo Clinic, Rochester, Minnesota, USA
| | | | - Joseph E. Parisi
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Tanis J. Ferman
- Department of Psychiatry and Psychology, Mayo Clinic, Jacksonville, FL, USA
| | | | | | | | | | - John D. Fryer
- Department of Neuroscience, Mayo Clinic, Scottsdale, AZ 85259, USA
| | - Hu Wang
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229
| | - Xianlin Han
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229
| | - Carl Frieden
- Department of Biochemistry and Molecular Biophysics, Washington University, St. Louis, MO, USA
| | | | - Owen A. Ross
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
- Department of Clinical Genomics, Mayo Clinic, Jacksonville, Florida, USA
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| |
Collapse
|
23
|
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder that currently has no cure. The aged population is growing globally, creating an urgent need for more promising therapies for this debilitating disease. Much effort has been made in recent decades, and the field is highly dynamic, with numerous trials. The main focus of these trials includes disease modification and symptomatic treatment. Some have shown beneficial outcomes, while others have shown no significant benefits. Here, we cover the outcome of recently published AD clinical trials, as well as the mechanism of action of these therapeutical agents, to re-think drug development strategies and directions for future studies.
Collapse
|
24
|
Farmer BC, Williams HC, Devanney NA, Piron MA, Nation GK, Carter DJ, Walsh AE, Khanal R, Young LEA, Kluemper JC, Hernandez G, Allenger EJ, Mooney R, Golden LR, Smith CT, Brandon JA, Gupta VA, Kern PA, Gentry MS, Morganti JM, Sun RC, Johnson LA. APOΕ4 lowers energy expenditure in females and impairs glucose oxidation by increasing flux through aerobic glycolysis. Mol Neurodegener 2021; 16:62. [PMID: 34488832 PMCID: PMC8420022 DOI: 10.1186/s13024-021-00483-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 08/15/2021] [Indexed: 01/21/2023] Open
Abstract
Background Cerebral glucose hypometabolism is consistently observed in individuals with Alzheimer’s disease (AD), as well as in young cognitively normal carriers of the Ε4 allele of Apolipoprotein E (APOE), the strongest genetic predictor of late-onset AD. While this clinical feature has been described for over two decades, the mechanism underlying these changes in cerebral glucose metabolism remains a critical knowledge gap in the field. Methods Here, we undertook a multi-omic approach by combining single-cell RNA sequencing (scRNAseq) and stable isotope resolved metabolomics (SIRM) to define a metabolic rewiring across astrocytes, brain tissue, mice, and human subjects expressing APOE4. Results Single-cell analysis of brain tissue from mice expressing human APOE revealed E4-associated decreases in genes related to oxidative phosphorylation, particularly in astrocytes. This shift was confirmed on a metabolic level with isotopic tracing of 13C-glucose in E4 mice and astrocytes, which showed decreased pyruvate entry into the TCA cycle and increased lactate synthesis. Metabolic phenotyping of E4 astrocytes showed elevated glycolytic activity, decreased oxygen consumption, blunted oxidative flexibility, and a lower rate of glucose oxidation in the presence of lactate. Together, these cellular findings suggest an E4-associated increase in aerobic glycolysis (i.e. the Warburg effect). To test whether this phenomenon translated to APOE4 humans, we analyzed the plasma metabolome of young and middle-aged human participants with and without the Ε4 allele, and used indirect calorimetry to measure whole body oxygen consumption and energy expenditure. In line with data from E4-expressing female mice, a subgroup analysis revealed that young female E4 carriers showed a striking decrease in energy expenditure compared to non-carriers. This decrease in energy expenditure was primarily driven by a lower rate of oxygen consumption, and was exaggerated following a dietary glucose challenge. Further, the stunted oxygen consumption was accompanied by markedly increased lactate in the plasma of E4 carriers, and a pathway analysis of the plasma metabolome suggested an increase in aerobic glycolysis. Conclusions Together, these results suggest astrocyte, brain and system-level metabolic reprogramming in the presence of APOE4, a ‘Warburg like’ endophenotype that is observable in young females decades prior to clinically manifest AD. Supplementary Information The online version contains supplementary material available at 10.1186/s13024-021-00483-y.
Collapse
Affiliation(s)
- Brandon C Farmer
- Department of Physiology, University of Kentucky College of Medicine, UKMC/MS 609, 800 Rose Street, Lexington, KY, 40536, USA
| | - Holden C Williams
- Department of Physiology, University of Kentucky College of Medicine, UKMC/MS 609, 800 Rose Street, Lexington, KY, 40536, USA.,Sanders Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Nicholas A Devanney
- Department of Physiology, University of Kentucky College of Medicine, UKMC/MS 609, 800 Rose Street, Lexington, KY, 40536, USA.,Sanders Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Margaret A Piron
- Department of Physiology, University of Kentucky College of Medicine, UKMC/MS 609, 800 Rose Street, Lexington, KY, 40536, USA
| | - Grant K Nation
- Department of Physiology, University of Kentucky College of Medicine, UKMC/MS 609, 800 Rose Street, Lexington, KY, 40536, USA
| | - David J Carter
- Department of Physiology, University of Kentucky College of Medicine, UKMC/MS 609, 800 Rose Street, Lexington, KY, 40536, USA
| | - Adeline E Walsh
- Department of Physiology, University of Kentucky College of Medicine, UKMC/MS 609, 800 Rose Street, Lexington, KY, 40536, USA
| | - Rebika Khanal
- Department of Physiology, University of Kentucky College of Medicine, UKMC/MS 609, 800 Rose Street, Lexington, KY, 40536, USA
| | - Lyndsay E A Young
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, USA
| | - Jude C Kluemper
- Department of Physiology, University of Kentucky College of Medicine, UKMC/MS 609, 800 Rose Street, Lexington, KY, 40536, USA
| | - Gabriela Hernandez
- Department of Physiology, University of Kentucky College of Medicine, UKMC/MS 609, 800 Rose Street, Lexington, KY, 40536, USA
| | - Elizabeth J Allenger
- Department of Physiology, University of Kentucky College of Medicine, UKMC/MS 609, 800 Rose Street, Lexington, KY, 40536, USA
| | - Rachel Mooney
- Department of Physiology, University of Kentucky College of Medicine, UKMC/MS 609, 800 Rose Street, Lexington, KY, 40536, USA
| | - Lesley R Golden
- Department of Physiology, University of Kentucky College of Medicine, UKMC/MS 609, 800 Rose Street, Lexington, KY, 40536, USA
| | - Cathryn T Smith
- Department of Physiology, University of Kentucky College of Medicine, UKMC/MS 609, 800 Rose Street, Lexington, KY, 40536, USA
| | - J Anthony Brandon
- Department of Physiology, University of Kentucky College of Medicine, UKMC/MS 609, 800 Rose Street, Lexington, KY, 40536, USA
| | - Vedant A Gupta
- Gill Heart and Vascular Institute, University of Kentucky, Lexington, KY, USA
| | - Philip A Kern
- Center for Clinical and Translational Science, University of Kentucky College of Medicine, Lexington, KY, USA.,Department of Internal Medicine, Division of Endocrinology, University of Kentucky, Lexington, KY, USA
| | - Matthew S Gentry
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, USA
| | - Josh M Morganti
- Sanders Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY, USA.,Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Ramon C Sun
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Lance A Johnson
- Department of Physiology, University of Kentucky College of Medicine, UKMC/MS 609, 800 Rose Street, Lexington, KY, 40536, USA. .,Sanders Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY, USA.
| |
Collapse
|
25
|
Wu D, Hu Y, Song M, Li G. Dichlorodiphenyltrichloroethane Impairs Amyloid Beta Clearance by Decreasing Liver X Receptor α Expression. Front Aging Neurosci 2021; 13:634948. [PMID: 34045954 PMCID: PMC8144330 DOI: 10.3389/fnagi.2021.634948] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 03/22/2021] [Indexed: 11/30/2022] Open
Abstract
Abnormal amyloid beta (Aβ) clearance is a distinctive pathological mechanism for Alzheimer’s disease (AD). ATP-binding cassette transporter A1 (ABCA1), which mediates the lipidation of apolipoprotein E, plays a critical role in Aβ clearance. As an environmental factor for AD, dichlorodiphenyltrichloroethane (DDT) can decrease ATP-binding cassette transporter A1 (ABCA1) expression and disrupt Aβ clearance. Liver X receptor α (LXRα) is an autoregulatory transcription factor for ABCA1 and a target of some environmental pollutants, such as organophosphate pesticides. In this study, we aimed to investigate whether DDT could affect Aβ clearance by targeting LXRα. The DDT-pretreated H4 human neuroglioma cells and immortalized astrocytes were incubated with exogenous Aβ to evaluate Aβ consumption. Meanwhile, cytotoxicity and LXRα expression were determined in the DDT-treated cells. Subsequently, the antagonism of DDT on LXRα agonist T0901317 was determined in vitro. The interaction between DDT and LXRα was predicted by molecular docking and molecular dynamics simulation technology. We observed that DDT could inhibit Aβ clearance and decrease the levels of LXRα mRNA and LXRα protein. Moreover, DDT is supposed to strongly bind to LXRα and exert antagonistic effects on LXRα. In conclusion, this study firstly presented that DDT could inhibit LXRα expression, which would contribute to Aβ clearance decline in vitro. It provides an experimental basis to search for potential therapeutic targets of AD.
Collapse
Affiliation(s)
- Dongmei Wu
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yang Hu
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Min Song
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Gongbo Li
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
26
|
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
|
27
|
Mallick S, Marshall PA, Wagner CE, Heck MC, Sabir ZL, Sabir MS, Dussik CM, Grozic A, Kaneko I, Jurutka PW. Evaluating Novel RXR Agonists That Induce ApoE and Tyrosine Hydroxylase in Cultured Human Glioblastoma Cells. ACS Chem Neurosci 2021; 12:857-871. [PMID: 33570383 DOI: 10.1021/acschemneuro.0c00707] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
There is considerable interest in identifying effective and safe drugs for neurodegenerative disorders. Cell culture and animal model work have demonstrated that modulating gene expression through RXR-mediated pathways may mitigate or reverse cognitive decline. However, because RXR is a dimeric partner for several transcription factors, activating off-target transcription is a concern with RXR ligands (rexinoids). This off-target gene modulation leads to unwanted side effects that can include low thyroid function and significant hyperlipidemia. There is a need to develop rexinoids that have binding specificity for subsets of RXR heterodimers, to drive desired gene modulation, but that do not induce spurious effects. Herein, we describe experiments in which we analyze a series of novel and previously reported rexinoids for their ability to modulate specific gene pathways implicated in neurodegenerative disorders employing a U87 cell culture model. We demonstrate that, compared to the FDA-approved rexinoid bexarotene (1), several of these compounds are equally or more effective at stimulating gene expression via LXREs or Nurr1/NBREs and are superior at inducing ApoE and/or tyrosine hydroxylase (TH) gene and protein expression, including analogs 8, 9, 13, 14, 20, 23, and 24, suggesting a possible therapeutic role for these compounds in Alzheimer's or Parkinson's disease (PD). A subset of these potent RXR agonists can synergize with a presumed Nurr1 ligand and antimalarial drug (amodiaquine) to further enhance Nurr1/NBREs-directed transcription. This novel discovery has potential clinical implications for treatment of PD since it suggests that the combination of an RXR agonist and a Nurr1 ligand can significantly enhance RXR-Nurr1 heterodimer activity and drive enhanced therapeutic expression of the TH gene to increase endogenous synthesis of dopamine. These data indicate that is it possible and prudent to develop novel rexinoids for testing of gene expression and side effect profiles for use in potential treatment of neurodegenerative disorders, as individual rexinoids can have markedly different gene expression profiles but similar structures.
Collapse
Affiliation(s)
- Sanchita Mallick
- School of Mathematical and Natural Sciences, Arizona State University, Phoenix, Arizona 85306, United States
| | - Pamela A. Marshall
- School of Mathematical and Natural Sciences, Arizona State University, Phoenix, Arizona 85306, United States
| | - Carl E. Wagner
- School of Mathematical and Natural Sciences, Arizona State University, Phoenix, Arizona 85306, United States
| | - Michael C. Heck
- School of Mathematical and Natural Sciences, Arizona State University, Phoenix, Arizona 85306, United States
| | - Zhela L. Sabir
- School of Mathematical and Natural Sciences, Arizona State University, Phoenix, Arizona 85306, United States
| | - Marya S. Sabir
- School of Mathematical and Natural Sciences, Arizona State University, Phoenix, Arizona 85306, United States
| | - Christoper M. Dussik
- School of Mathematical and Natural Sciences, Arizona State University, Phoenix, Arizona 85306, United States
| | - Aleksandra Grozic
- School of Mathematical and Natural Sciences, Arizona State University, Phoenix, Arizona 85306, United States
| | - Ichiro Kaneko
- School of Mathematical and Natural Sciences, Arizona State University, Phoenix, Arizona 85306, United States
| | - Peter W. Jurutka
- School of Mathematical and Natural Sciences, Arizona State University, Phoenix, Arizona 85306, United States
| |
Collapse
|
28
|
Jonathan MC, Adrián SH, Gonzalo A. Type II nuclear receptors with potential role in Alzheimer disease. Mol Aspects Med 2021; 78:100940. [PMID: 33397589 DOI: 10.1016/j.mam.2020.100940] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 02/06/2023]
Abstract
Nuclear receptors are ligand-activated transcription factors that can modulated cellular processes involved in the development, homeostasis, cell proliferation, metabolism, and reproduction through the control of the specific genetic and molecular program. In the central nervous system, they are key regulators of neural stem cell fate decisions and can modulate the physiology of different brain cells. Over the past decades, a large body of evidence has supported that nuclear receptors are potential therapeutic targets for the treatment of neurodegenerative disorders such as Alzheimer's disease, the most common dementia worldwide, and the main cause of disability in later life. This disease is characterized by the progressive accumulation of amyloid-beta peptides and hyperphosphorylated tau protein that can explain alterations in synaptic transmission and plasticity; loss of dendritic spines; increased in reactive microglia and inflammation; reduction of neuronal stem cells number; myelin and vascular alterations that finally leads to increased neuronal death. Here, we present a review of type II no steroidal nuclear receptors that form obligatory heterodimers with the Retinoid X Receptor (RXR) and its potential in the therapeutic of AD. Activation of type II nuclear receptor by synthetic agonist leads to transcriptional regulation of specific genes that acts counteracting against the detrimental effects of amyloid-beta peptides and hyperphosphorylated tau in neuronal cells recovering the functionality of the synapses. But also, activation of type II nuclear receptor leads to modifications in APP metabolism, repression of inflammatory cascade and inductors of the generation of neuronal stem cells and progenitor cells supporting its potential therapeutics role for Alzheimer's disease.
Collapse
Affiliation(s)
- Muñoz-Cabrera Jonathan
- Grupo de Neurociencias y Muerte Celular, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Sandoval-Hernández Adrián
- Grupo de Neurociencias y Muerte Celular, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá, Colombia; Departamento de Química, Facultad de Ciencias, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Arboleda Gonzalo
- Grupo de Neurociencias y Muerte Celular, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá, Colombia; Departamento de Patología, Facultad de Medicina, Universidad Nacional de Colombia, Bogotá, Colombia.
| |
Collapse
|
29
|
Effects of 2,4-thiazolidinedione (TZD) on milk fatty acid profile and serum vitamins in dairy goats challenged with intramammary infusion of Streptococcus uberis. J DAIRY RES 2020; 87:416-423. [PMID: 33168108 DOI: 10.1017/s0022029920000941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The study included two experiments. In the first, 24 lactating Saanen dairy goats received low-energy diet without vitamin supplements. Twelve goats received a daily IV injection of 2,4- thiazolidinedione (TZD), others received saline injection. A week later, 6 goats from each treatment were challenged with intramammary infusion (IMI) of saline (CTRL) or Streptococcus uberis. In the second experiment, 12 Saanen lactating dairy goats received supplemental vitamins to reach NRC recommendation level. Six goats in each group were injected with TZD or saline daily, and 14 d later received Streptococcus uberis IMI in the right half of the udder. The hypotheses were (1) TZD does not affect the level of retinol in blood, and (2) the fatty acid profile is affected by the interaction between mammary infection and TZD in dairy goats. In the first experiment blood samples were collected on d -7, -2, 1, 2, 12 and milk samples were collected on d -8, 1, 4, 7, and 12, both relative to IMI. In the second experiment, blood samples were collected on d -15, 0, 1, and 10 relative to IMI. Milk and serum samples were analyzed for retinol, α-tocopherol and fatty acid profile. Serum retinol and β-carotene concentrations were higher in the second experiment compared to the first. Serum β-carotene and α-tocopherol were greater in TZD than CTRL and there was a TZD × time interaction in the first experiment. In addition, the TZD × time interaction showed that the milk fatty acid were reduced in C16 : 0 while C18 : 3 n3 while total omega 3 fatty acids were increased, as well as with minor effect on preventing a transient increase in α-tocopherol in milk. Overall, the TZD may affect the lipid-soluble vitamins and fatty acid profile, potentially altering immune responses, during mastitis in dairy goats.
Collapse
|
30
|
Bicker J, Alves G, Fonseca C, Falcão A, Fortuna A. Repairing blood-CNS barriers: Future therapeutic approaches for neuropsychiatric disorders. Pharmacol Res 2020; 162:105226. [PMID: 33007420 DOI: 10.1016/j.phrs.2020.105226] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 09/21/2020] [Accepted: 09/22/2020] [Indexed: 12/19/2022]
Abstract
Central nervous system (CNS) drug development faces significant difficulties that translate into high rates of failure and lack of innovation. The pathophysiology of neurological and psychiatric disorders often results in the breakdown of blood-CNS barriers, disturbing the CNS microenvironment and worsening disease progression. Therefore, restoring the integrity of blood-CNS barriers may have a beneficial influence in several CNS disorders and improve treatment outcomes. In this review, pathways that may be modulated to protect blood-CNS barriers from neuroinflammatory and oxidative insults are featured. First, the participation of the brain endothelium and glial cells in disruption processes is discussed. Then, the relevance of regulatory systems is analysed, specifically the hypothalamic-pituitary axis, the renin-angiotensin system, sleep and circadian rhythms, and glutamate neurotransmission. Lastly, compounds of endogenous and exogenous origin that are known to mediate the repair of blood-CNS barriers are presented. We believe that enhancing the protection of blood-CNS barriers is a promising therapeutic strategy to pursue in the future.
Collapse
Affiliation(s)
- Joana Bicker
- University of Coimbra, Faculty of Pharmacy, Coimbra, Portugal; University of Coimbra, Coimbra Institute for Biomedical Imaging and Translational Research, Coimbra, Portugal.
| | - Gilberto Alves
- CICS-UBI, Health Sciences Research Center, University of Beira Interior, Covilhã, Portugal
| | - Carla Fonseca
- University of Coimbra, Faculty of Pharmacy, Coimbra, Portugal
| | - Amílcar Falcão
- University of Coimbra, Faculty of Pharmacy, Coimbra, Portugal; University of Coimbra, Coimbra Institute for Biomedical Imaging and Translational Research, Coimbra, Portugal
| | - Ana Fortuna
- University of Coimbra, Faculty of Pharmacy, Coimbra, Portugal; University of Coimbra, Coimbra Institute for Biomedical Imaging and Translational Research, Coimbra, Portugal
| |
Collapse
|
31
|
Clark JN, Whiting A, McCaffery P. Retinoic acid receptor-targeted drugs in neurodegenerative disease. Expert Opin Drug Metab Toxicol 2020; 16:1097-1108. [DOI: 10.1080/17425255.2020.1811232] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Jason Nicol Clark
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| | | | - Peter McCaffery
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| |
Collapse
|
32
|
Plascencia-Villa G, Perry G. Status and future directions of clinical trials in Alzheimer's disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 154:3-50. [PMID: 32739008 DOI: 10.1016/bs.irn.2020.03.022] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Amyloid-β (Aβ) senile plaques and neurofibrillary tangles of tau are generally recognized as the culprits of Alzheimer's disease (AD) and related dementia. About 25 years ago, the amyloid cascade hypotheses postulated a direct correlation of plaques with the development of AD, and it has been the dominant theory since then. In this period, more than 200 clinical trials focused mainly on targeting components of the Aβ cascade have dramatically failed, some of them in Phase III. With a greater than 99.6% failure rate at a cost of several billion from governments, industry, and private funders, therapeutic strategies targeting amyloid and tau are now under scrutiny. Therefore, it is time to reevaluate alternatives to targeting Aβ and tau as effective therapeutic strategies for AD. The diagnosis of AD is currently based on medical examination of symptoms including tests to assess memory impairment, attention, language, and other thinking skills. This is complemented with brain scans, such as computed tomography, magnetic resonance imaging, or positron emission tomography with the help of imaging probes targeting Aβ or tau deposits. This approach has contributed to the tunnel vision focus on Aβ and tau as the main culprits of AD. However, events upstream of these proteopathies (age-related impaired neuronal bioenergetics, lysosome function, neurotrophic signaling, and neuroinflammation, among others) are almost surely where the development of alternative therapeutic interventions should be targeted. Here, we present the current status of therapeutic candidates targeting diverse mechanisms and strategies including Aβ and tau, proteins involved in Aβ production and trafficking (ApoE, α/β/γ-secretases), neuroinflammation, neurotransmitters, neuroprotective agents antimicrobials, and gene and stem cell therapy. There are currently around 33 compounds in Phase III, 78 in Phase II, and 32 more in Phase I trials. With the current world health crisis of increased dementia in a rapidly aging population, effective AD therapies are desperately needed.
Collapse
Affiliation(s)
- Germán Plascencia-Villa
- Department of Biology and Neurosciences Institute, The University of Texas at San Antonio (UTSA), 1 UTSA Circle, San Antonio, TX, United States
| | - George Perry
- Department of Biology and Neurosciences Institute, The University of Texas at San Antonio (UTSA), 1 UTSA Circle, San Antonio, TX, United States.
| |
Collapse
|
33
|
Sáez-Orellana F, Octave JN, Pierrot N. Alzheimer's Disease, a Lipid Story: Involvement of Peroxisome Proliferator-Activated Receptor α. Cells 2020; 9:E1215. [PMID: 32422896 PMCID: PMC7290654 DOI: 10.3390/cells9051215] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/10/2020] [Accepted: 05/12/2020] [Indexed: 12/11/2022] Open
Abstract
Alzheimer's disease (AD) is the leading cause of dementia in the elderly. Mutations in genes encoding proteins involved in amyloid-β peptide (Aβ) production are responsible for inherited AD cases. The amyloid cascade hypothesis was proposed to explain the pathogeny. Despite the fact that Aβ is considered as the main culprit of the pathology, most clinical trials focusing on Aβ failed and suggested that earlier interventions are needed to influence the course of AD. Therefore, identifying risk factors that predispose to AD is crucial. Among them, the epsilon 4 allele of the apolipoprotein E gene that encodes the major brain lipid carrier and metabolic disorders such as obesity and type 2 diabetes were identified as AD risk factors, suggesting that abnormal lipid metabolism could influence the progression of the disease. Among lipids, fatty acids (FAs) play a fundamental role in proper brain function, including memory. Peroxisome proliferator-activated receptor α (PPARα) is a master metabolic regulator that regulates the catabolism of FA. Several studies report an essential role of PPARα in neuronal function governing synaptic plasticity and cognition. In this review, we explore the implication of lipid metabolism in AD, with a special focus on PPARα and its potential role in AD therapy.
Collapse
Affiliation(s)
- Francisco Sáez-Orellana
- Université Catholique de Louvain, Alzheimer Dementia, Avenue Mounier 53, SSS/IONS/CEMO-Bte B1.53.03, B-1200 Brussels, Belgium; (F.S.-O.); (J.-N.O.)
- Institute of Neuroscience, Alzheimer Dementia, Avenue Mounier 53, SSS/IONS/CEMO-Bte B1.53.03, B-1200 Brussels, Belgium
| | - Jean-Noël Octave
- Université Catholique de Louvain, Alzheimer Dementia, Avenue Mounier 53, SSS/IONS/CEMO-Bte B1.53.03, B-1200 Brussels, Belgium; (F.S.-O.); (J.-N.O.)
- Institute of Neuroscience, Alzheimer Dementia, Avenue Mounier 53, SSS/IONS/CEMO-Bte B1.53.03, B-1200 Brussels, Belgium
| | - Nathalie Pierrot
- Université Catholique de Louvain, Alzheimer Dementia, Avenue Mounier 53, SSS/IONS/CEMO-Bte B1.53.03, B-1200 Brussels, Belgium; (F.S.-O.); (J.-N.O.)
- Institute of Neuroscience, Alzheimer Dementia, Avenue Mounier 53, SSS/IONS/CEMO-Bte B1.53.03, B-1200 Brussels, Belgium
| |
Collapse
|
34
|
Zhao H, Li S, Li Z, Yang S, Li D, Zheng J, Gao H, Yun L, Gu Y, Li L, Zhao J, Fu Y. Intranasal delivery of 9-cis retinoic acid reduces beta-amyloid deposition via inhibiting astrocyte-mediated inflammation. Aging (Albany NY) 2020; 12:5469-5478. [PMID: 32209731 PMCID: PMC7138573 DOI: 10.18632/aging.102970] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Accepted: 01/27/2020] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) is associated with the accumulation and deposition of a beta-amyloid (Αβ) peptide in the brain, resulting in increased neuroinflammation and synaptic dysfunction. Intranasal delivery of targeted drugs to the brain represents a noninvasive pathway that bypasses the blood-brain barrier and minimizes systemic exposure. The aim of this study was to evaluate the therapeutic effect of intranasally delivered 9-cis retinoic acid (RA) on the neuropathology of an AD mouse model. Herein, we observed dramatically decreased Αβ deposition in the brains of amyloid precursor protein (APP) and presenilin 1 (PS1) double-transgenic mice (APP/PS1) treated intranasally with 9-cis RA for 4 weeks compared to that in the brains of vehicle-treated mice. Importantly, intranasal delivery of 9-cis RA suppressed Αβ-associated astrocyte activation and neuroinflammation and ultimately restored synaptic deficits in APP/PS1 transgenic mice. These results support the critical roles of Αβ-associated neuroinflammation responses to synaptic deficits, particularly during the deposition of Αβ. Our findings provide strong evidence that intranasally delivered 9-cis RA attenuates neuronal dysfunction in an AD mouse model and is a promising therapeutic strategy for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Hong Zhao
- Department of Neurology, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Shuo Li
- Department of Ultrasonography, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Zhuo Li
- Department of Endocrinology, The First Hospital of Jilin University, Changchun, China
| | - Shuo Yang
- Department of Neurology, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Dandan Li
- Department of Neurology, The Second Hospital of Heilongjiang Province, Harbin, China
| | - Jiaolin Zheng
- Department of Neurology, The Second Hospital of Harbin Medical University, Harbin, China
| | - Hongmei Gao
- Department of Neurology, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Ling Yun
- Department of Neurology, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - YingLi Gu
- Department of Neurology, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Longxuan Li
- Department of Neurology, Gongli Hospital of The Second Military Medical University, Shanghai, China
| | - Jing Zhao
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Yuan Fu
- Department of Neurology, The Fourth Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
35
|
Chang CF, Massey J, Osherov A, Angenendt da Costa LH, Sansing LH. Bexarotene Enhances Macrophage Erythrophagocytosis and Hematoma Clearance in Experimental Intracerebral Hemorrhage. Stroke 2019; 51:612-618. [PMID: 31826730 DOI: 10.1161/strokeaha.119.027037] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Background and Purpose- Enhancement of erythrophagocytosis by macrophages in a timely manner can limit the toxic effects of erythrocyte metabolites and promote brain recovery after intracerebral hemorrhage (ICH). In the current study, we investigated the therapeutic effect of retinoid X receptor agonist, bexarotene, in facilitating erythrophagocytosis and neurobehavioral recovery in 2 mouse models of ICH. Methods- Bone marrow-derived macrophages and fluorescently labeled erythrocytes were used to study erythrophagocytosis in vitro with phenotypic changes quantified by gene expression. ICH was modeled in vivo using intrastriatal autologous blood and collagenase injection in mice with and without bexarotene treatment beginning 3 hours after ICH. In vivo phagocytosis, ability and hematoma clearance were evaluated by erythrophagocytosis assays, flow cytometry, and histological analysis. Neurological deficits and functional recovery were also quantified. Results- Bexarotene increased macrophage expression of phagocytosis receptors and erythrophagocytosis and reduced macrophage TNF (tumor necrosis factor) production in vitro. In vivo, bexarotene treatment enhanced erythrophagocytosis, reduced hematoma volume, and ultimately improved neurological recovery after ICH in 2 distinct models of ICH. Conclusions- Bexarotene administration is beneficial for recovery after ICH by enhancing hemorrhage phagocytosis, modulating macrophage phenotype, and improving functional recovery.
Collapse
Affiliation(s)
- Che-Feng Chang
- From the Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei (C.-F.C.).,Department of Neurology, Yale University School of Medicine, New Haven, CT (C.-F.C., J.M., A.O., L.H.S.)
| | - Jordan Massey
- Department of Neurology, Yale University School of Medicine, New Haven, CT (C.-F.C., J.M., A.O., L.H.S.)
| | - Artem Osherov
- Department of Neurology, Yale University School of Medicine, New Haven, CT (C.-F.C., J.M., A.O., L.H.S.)
| | - Luís Henrique Angenendt da Costa
- Department of Neurosciences and Behavioral Sciences, Ribeirao Preto Medical School, University of Sao Paulo, Brazil (L.H.A.d.C.)
| | - Lauren H Sansing
- Department of Neurology, Yale University School of Medicine, New Haven, CT (C.-F.C., J.M., A.O., L.H.S.)
| |
Collapse
|
36
|
Patra K, Giannisis A, Edlund AK, Sando SB, Lauridsen C, Berge G, Grøntvedt GR, Bråthen G, White LR, Nielsen HM. Plasma Apolipoprotein E Monomer and Dimer Profile and Relevance to Alzheimer's Disease. J Alzheimers Dis 2019; 71:1217-1231. [PMID: 31524156 DOI: 10.3233/jad-190175] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The APOEɛ4 gene variant is the strongest genetic risk factor for Alzheimer's disease (AD), whereas APOEɛ3 conventionally is considered as 'risk neutral' although APOEɛ3-carriers also develop AD. Previous studies have shown that the apolipoprotein E3 (apoE3) isoform occurs as monomers, homodimers and heterodimers with apolipoprotein A-II in human body fluids and brain tissue, but the relevance of a plasma apoE3 monomer/dimer profile to AD is unknown. Here we assessed the distribution of monomers, homodimers and heterodimers in plasma from control subjects and patients with mild cognitive impairment (MCI) and AD with either a homozygous APOEɛ3 (n = 31 control subjects, and n = 14 MCI versus n = 5 AD patients) or APOEɛ4 genotype (n = 1 control subject, n = 21 MCI and n = 7 AD patients). Total plasma apoE levels were lower in APOEɛ4-carriers and overall correlated significantly to CSF Aβ42, p(Thr181)-tau and t-tau levels. Apolipoprotein E dimers were only observed in the APOEɛ3-carriers and associated with total plasma apoE levels, negatively correlated to apoE monomers, but were unrelated to plasma homocysteine levels. Importantly, the APOEɛ3-carrying AD patients versus controls exhibited a significant decrease in apoE homodimers (17.8±9.6% versus 26.7±6.3%, p = 0.025) paralleled by an increase in apoE monomers (67.8±18.3% versus 48.5±11.2%, p = 0.008). In the controls, apoE monomers and heterodimers were significantly associated with plasma triglycerides; the apoE heterodimers were also associated with levels of high-density lipoprotein cholesterol. The physiological relevance of apoE dimer formation needs to be further investigated, though the distribution of apoE in monomers and dimers appears to be of relevance to AD in APOEɛ3 subjects.
Collapse
Affiliation(s)
- Kalicharan Patra
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Andreas Giannisis
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Anna K Edlund
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Sigrid Botne Sando
- Department of Neurology, University Hospital of Trondheim, Trondheim, Norway.,Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology, Trondheim, Norway
| | - Camilla Lauridsen
- Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology, Trondheim, Norway
| | - Guro Berge
- Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology, Trondheim, Norway
| | | | - Geir Bråthen
- Department of Neurology, University Hospital of Trondheim, Trondheim, Norway.,Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology, Trondheim, Norway
| | - Linda R White
- Department of Neurology, University Hospital of Trondheim, Trondheim, Norway.,Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology, Trondheim, Norway
| | - Henrietta M Nielsen
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| |
Collapse
|
37
|
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
|
38
|
Shinohara M, Shinohara M, Zhao J, Fu Y, Liu CC, Kanekiyo T, Bu G. 5-HT3 Antagonist Ondansetron Increases apoE Secretion by Modulating the LXR-ABCA1 Pathway. Int J Mol Sci 2019; 20:ijms20061488. [PMID: 30934555 PMCID: PMC6471172 DOI: 10.3390/ijms20061488] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 03/10/2019] [Accepted: 03/21/2019] [Indexed: 12/20/2022] Open
Abstract
Apolipoprotein E (apoE) is linked to the risk for Alzheimer’s disease (AD) and thus has been suggested to be an important therapeutic target. In our drug screening effort, we identified Ondansetron (OS), an FDA-approved 5-HT3 antagonist, as an apoE-modulating drug. OS at low micromolar concentrations significantly increased apoE secretion from immortalized astrocytes and primary astrocytes derived from apoE3 and apoE4-targeted replacement mice without generating cellular toxicity. Other 5-HT3 antagonists also had similar effects as OS, though their effects were milder and required higher concentrations. Antagonists for other 5-HT receptors did not increase apoE secretion. OS also increased mRNA and protein levels of the ATB-binding cassette protein A1 (ABCA1), which is involved in lipidation and secretion of apoE. Accordingly, OS increased high molecular weight apoE. Moreover, the liver X receptor (LXR) and ABCA1 antagonists blocked the OS-induced increase of apoE secretion, indicating that the LXR-ABCA1 pathway is involved in the OS-mediated facilitation of apoE secretion from astrocytes. The effects of OS on apoE and ABCA1 were also observed in human astrocytes derived from induced pluripotent stem cells (iPSC) carrying the APOE ε3/ε3 and APOE ε4/ε4 genotypes. Oral administration of OS at clinically-relevant doses affected apoE levels in the liver, though the effects in the brain were not observed. Collectively, though further studies are needed to probe its effects in vivo, OS could be a potential therapeutic drug for AD by modulating poE metabolism through the LXR-ABCA1 pathway.
Collapse
Affiliation(s)
- Motoko Shinohara
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA.
- Department of Aging Neurobiology, National Center for Geriatrics and Gerontology, Aichi 474-8511, Japan.
| | - Mitsuru Shinohara
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA.
- Department of Aging Neurobiology, National Center for Geriatrics and Gerontology, Aichi 474-8511, Japan.
| | - Jing Zhao
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA.
| | - Yuan Fu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA.
| | - Chia-Chen Liu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA.
| | - Takahisa Kanekiyo
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA.
- Neuroscience Graduate Program, Mayo Clinic, Jacksonville, FL 32224, USA.
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA.
- Neuroscience Graduate Program, Mayo Clinic, Jacksonville, FL 32224, USA.
| |
Collapse
|
39
|
Farmer BC, Kluemper J, Johnson LA. Apolipoprotein E4 Alters Astrocyte Fatty Acid Metabolism and Lipid Droplet Formation. Cells 2019; 8:cells8020182. [PMID: 30791549 PMCID: PMC6406677 DOI: 10.3390/cells8020182] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 02/15/2019] [Accepted: 02/19/2019] [Indexed: 12/11/2022] Open
Abstract
Lipid droplets (LDs) serve as energy rich reservoirs and have been associated with apolipoprotein E (APOE) and neurodegeneration. The E4 allele of APOE (E4) is the strongest genetic risk factor for the development of late onset Alzheimer's disease (AD). Since both E4 carriers and individuals with AD exhibit a state of cerebral lipid dyshomeostasis, we hypothesized that APOE may play a role in regulating LD metabolism. We found that astrocytes expressing E4 accumulate significantly more and smaller LDs compared to E3 astrocytes. Accordingly, expression of perilipin-2, an essential LD protein component, was higher in E4 astrocytes. We then probed fatty acid (FA) metabolism and found E4 astrocytes to exhibit decreased uptake of palmitate, and decreased oxidation of exogenously supplied oleate and palmitate. We then measured oxygen consumption rate, and found E4 astrocytes to consume more oxygen for endogenous FA oxidation and accumulate more LD-derived metabolites due to incomplete oxidation. Lastly, we found that E4 astrocytes are more sensitive to carnitine palmitoyltransferase-1 inhibition than E3 astrocytes. These findings offer the potential for further studies investigating the link between astrocyte lipid storage, utilization, and neurodegenerative disease as a function of APOE genotype.
Collapse
Affiliation(s)
- Brandon C Farmer
- Department of Physiology, University of Kentucky, 800 Rose Street, Rm: MS-609, Lexington, KY 40536, USA.
| | - Jude Kluemper
- Department of Physiology, University of Kentucky, 800 Rose Street, Rm: MS-609, Lexington, KY 40536, USA.
| | - Lance A Johnson
- Department of Physiology, University of Kentucky, 800 Rose Street, Rm: MS-609, Lexington, KY 40536, USA.
- Sanders-Brown Center on Aging, University of Kentucky, 800 Rose Street, Lexington, KY 40536, USA.
| |
Collapse
|
40
|
Clemens V, Regen F, Le Bret N, Heuser I, Hellmann-Regen J. Retinoic Acid Enhances Apolipoprotein E Synthesis in Human Macrophages. J Alzheimers Dis 2019; 61:1295-1300. [PMID: 29376871 DOI: 10.3233/jad-170823] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Apolipoprotein E (ApoE) represents a pivotal target in Alzheimer's disease (AD) and is modulated through retinoic acid (RA), an endogenous neuroprotective and anti-inflammatory compound. A major source of ApoE are microglia, which are pathologically activated in AD. Activated microglia are known to block RA signaling. This suggests a vicious cycle between inflammation, RA signaling, and ApoE homeostasis in AD pathogenesis. To test this hypothesis, we investigated effects of RA and proinflammatory activation on ApoE synthesis in primary human macrophage-derived microglial-like cells. Our results indicate that proinflammatory activation attenuates ApoE synthesis, an effect blocked by RA.
Collapse
Affiliation(s)
- Vera Clemens
- Department of Psychiatry, Section Clinical Neurobiology, Campus Benjamin Franklin, Charité, University Medicine Berlin, Germany
| | - Francesca Regen
- Department of Psychiatry, Section Clinical Neurobiology, Campus Benjamin Franklin, Charité, University Medicine Berlin, Germany
| | - Nathalie Le Bret
- Department of Psychiatry, Section Clinical Neurobiology, Campus Benjamin Franklin, Charité, University Medicine Berlin, Germany
| | - Isabella Heuser
- Department of Psychiatry, Section Clinical Neurobiology, Campus Benjamin Franklin, Charité, University Medicine Berlin, Germany
| | - Julian Hellmann-Regen
- Department of Psychiatry, Section Clinical Neurobiology, Campus Benjamin Franklin, Charité, University Medicine Berlin, Germany
| |
Collapse
|
41
|
He J, Liu H, Zhong J, Guo Z, Wu J, Zhang H, Huang Z, Jiang L, Li H, Zhang Z, Liu L, Wu Y, Qi L, Sun X, Cheng C. Bexarotene protects against neurotoxicity partially through a PPARγ-dependent mechanism in mice following traumatic brain injury. Neurobiol Dis 2018; 117:114-124. [DOI: 10.1016/j.nbd.2018.06.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 05/05/2018] [Accepted: 06/06/2018] [Indexed: 12/20/2022] Open
|
42
|
Shackleton B, Crawford F, Bachmeier C. Apolipoprotein E-mediated Modulation of ADAM10 in Alzheimer's Disease. Curr Alzheimer Res 2018; 14:578-585. [PMID: 28164773 DOI: 10.2174/1567205014666170203093219] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 01/19/2017] [Accepted: 01/31/2017] [Indexed: 11/22/2022]
Abstract
BACKGROUND The APOE4 allele is the strongest genetic risk factor for Alzheimer's disease (AD). It has been associated with an accumulation of amyloid-β (Aβ) in the brain, which is produced through the sequential cleavage of the amyloid-β precursor protein (AβPP) by β - and γ-secretases. Alternatively, AβPP is also cleaved by α -secretases such as A Disintegrin and Metalloproteinase Domain-containing Protein 10 (ADAM10). OBJECTIVE While several studies have investigated the impact of apoE on β- and γ-secretase, interactions between apoE and α-secretases have not been fully examined. We investigated the effect of each apoE isoform on ADAM10 in vitro and in human cortex samples. METHOD ADAM10 activity and kinetics was assessed in cell-free assays and the biological activity of ADAM10 further investigated in 7WCHO cells over-expressing wild type AβPP through ELISA. Finally, ADAM10 expression and activity was observed in the soluble fraction of both control and Alzheimer's Disease human cortex samples through ELISA. RESULTS In a cell free assay, ADAM10 activity was found to be significantly lower in apoE4 samples compared to apoE2. 7WCHO cells over expressing wild type AβPP exposed to apoE4 demonstrated reduced formation of sAβPPα compared to other apoE isoforms. We also identified APOE and AD dependent changes in ADAM10 activity and expression in the soluble brain fraction of human brain cortex. CONCLUSION Overall, our data demonstrates an apoE isoform-dependent effect on ADAM10 function and AβPP processing which may describe the elevated amyloid levels in the brains of AD subjects carrying the APOE4 allele.
Collapse
Affiliation(s)
- Ben Shackleton
- 2040 Whitfield Avenue, Sarasota, Florida 34243, United States
| | - Fiona Crawford
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, Florida, 34243, United States
| | - Corbin Bachmeier
- The Open University, Walton Hall, Milton Keynes, Buckinghamshire, MK7 6AA, United Kingdom
| |
Collapse
|
43
|
Liu CC, Zhao N, Fu Y, Wang N, Linares C, Tsai CW, Bu G. ApoE4 Accelerates Early Seeding of Amyloid Pathology. Neuron 2017; 96:1024-1032.e3. [PMID: 29216449 DOI: 10.1016/j.neuron.2017.11.013] [Citation(s) in RCA: 222] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 09/08/2017] [Accepted: 11/09/2017] [Indexed: 12/29/2022]
Abstract
Accumulation and aggregation of amyloid-β (Aβ) in the brain is an initiating step in the pathogenesis of Alzheimer's disease (AD). The ε4 allele of apolipoprotein E (apoE) gene is the strongest genetic risk factor for late-onset AD. Although there is strong evidence showing that apoE4 enhances amyloid pathology, it is not clear what the critical stage(s) is during amyloid development in which apoE4 has the strongest impact. Using apoE inducible mouse models, we show that increased expression of astrocytic apoE4, but not apoE3, during the seeding stage of amyloid development enhanced amyloid deposition and neuritic dystrophy in amyloid model mice. ApoE4, but not apoE3, significantly increased brain Aβ half-life measured by in vivo microdialysis. Furthermore, apoE4 expression increased whereas apoE3 reduced amyloid-related gliosis in the mouse brains. Together, our results demonstrate that apoE4 has the greatest impact on amyloid during the seeding stage, likely by perturbing Aβ clearance and enhancing Aβ aggregation.
Collapse
Affiliation(s)
- Chia-Chen Liu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA.
| | - Na Zhao
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Yuan Fu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Na Wang
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA; Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, Fujian 361005, China
| | - Cynthia Linares
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Chih-Wei Tsai
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA; Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, College of Medicine, Xiamen University, Xiamen, Fujian 361005, China.
| |
Collapse
|
44
|
Di Battista AM, Heinsinger NM, Rebeck GW. Alzheimer's Disease Genetic Risk Factor APOE-ε4 Also Affects Normal Brain Function. Curr Alzheimer Res 2017; 13:1200-1207. [PMID: 27033053 DOI: 10.2174/1567205013666160401115127] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 03/15/2016] [Accepted: 03/28/2016] [Indexed: 12/24/2022]
Abstract
APOE-ε4 is the strongest genetic risk factor for Alzheimer's disease (AD), and is associated with an increase in the levels of amyloid deposition and an early age of onset. Recent data demonstrate that AD pathological changes occur decades before clinical symptoms, raising questions about the precise onset of the disease. Now a convergence of approaches in mice and humans has demonstrated that APOE-ε4 affects normal brain function even very early in life in the absence of gross AD pathological changes. Normal mice expressing APOE4 have task-specific spatial learning deficits, as well as reduced NMDAR-dependent signaling and structural changes to presynaptic and postsynaptic compartments in neurons, particularly in hippocampal regions. Young humans possessing APOE-ε4 are more adept than APOE-ε4 negative individuals at some behavioral tasks, and functional magnetic resonance imaging has shown that inheritance of APOE-ε4 has specific effects on medial temporal brain activities. These findings suggest that inheritance of APOE-ε4 causes life long changes to the brain that may be related to the late risk of AD. Several possible mechanisms of how APOE-ε4 could affect brain neurochemistry, structure, and function are reviewed.
Collapse
Affiliation(s)
| | | | - G William Rebeck
- New Research Building, WP- 13, 3970 Reservoir Rd, NW, Washington, DC 20007; USA
| |
Collapse
|
45
|
Yang DS, Stavrides P, Kumar A, Jiang Y, Mohan PS, Ohno M, Dobrenis K, Davidson CD, Saito M, Pawlik M, Huo C, Walkley SU, Nixon RA. Cyclodextrin has conflicting actions on autophagy flux in vivo in brains of normal and Alzheimer model mice. Hum Mol Genet 2017; 26:843-859. [PMID: 28062666 DOI: 10.1093/hmg/ddx001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 01/03/2016] [Indexed: 12/13/2022] Open
Abstract
2-hydroxypropyl-β-cyclodextrin (CYCLO), a modifier of cholesterol efflux from cellular membrane and endo-lysosomal compartments, reduces lysosomal lipid accumulations and has therapeutic effects in animal models of Niemann-Pick disease type C and several other neurodegenerative states. Here, we investigated CYCLO effects on autophagy in wild-type mice and TgCRND8 mice-an Alzheimer's Disease (AD) model exhibiting β-amyloidosis, neuronal autophagy deficits leading to protein and lipid accumulation within greatly enlarged autolysosomes. A 14-day intracerebroventricular administration of CYCLO to 8-month-old TgCRND8 mice that exhibit moderately advanced neuropathology markedly diminished the sizes of enlarged autolysosomes and lowered their content of GM2 ganglioside and Aβ-immunoreactivity without detectably altering amyloid precursor protein processing or extracellular Aβ/β-amyloid burden. We identified two major actions of CYCLO on autophagy underlying amelioration of lysosomal pathology. First, CYCLO stimulated lysosomal proteolytic activity by increasing cathepsin D activity, levels of cathepsins B and D and two proteins known to interact with cathepsin D, NPC1 and ABCA1. Second, CYCLO impeded autophagosome-lysosome fusion as evidenced by the accumulation of LC3, SQSTM1/p62, and ubiquitinated substrates in an expanded population of autophagosomes in the absence of greater autophagy induction. By slowing substrate delivery to lysosomes, autophagosome maturational delay, as further confirmed by our in vitro studies, may relieve lysosomal stress due to accumulated substrates. These findings provide in vivo evidence for lysosomal enhancing properties of CYCLO, but caution that prolonged interference with cellular membrane fusion/autophagosome maturation could have unfavorable consequences, which might require careful optimization of dosage and dosing schedules.
Collapse
Affiliation(s)
- Dun-Sheng Yang
- Nathan Kline Institute, Orangeburg, NY, USA.,Department of Psychiatry, New York University Langone Medical Center, New York, NY, USA
| | | | - Asok Kumar
- Nathan Kline Institute, Orangeburg, NY, USA.,Department of Psychiatry, New York University Langone Medical Center, New York, NY, USA
| | - Ying Jiang
- Nathan Kline Institute, Orangeburg, NY, USA.,Department of Psychiatry, New York University Langone Medical Center, New York, NY, USA
| | - Panaiyur S Mohan
- Nathan Kline Institute, Orangeburg, NY, USA.,Department of Psychiatry, New York University Langone Medical Center, New York, NY, USA
| | - Masuo Ohno
- Nathan Kline Institute, Orangeburg, NY, USA.,Department of Psychiatry, New York University Langone Medical Center, New York, NY, USA
| | - Kostantin Dobrenis
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Cristin D Davidson
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Mitsuo Saito
- Nathan Kline Institute, Orangeburg, NY, USA.,Department of Psychiatry, New York University Langone Medical Center, New York, NY, USA
| | | | | | - Steven U Walkley
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ralph A Nixon
- Nathan Kline Institute, Orangeburg, NY, USA.,Department of Psychiatry, New York University Langone Medical Center, New York, NY, USA.,Cell Biology, New York University Langone Medical Center, New York, NY, USA
| |
Collapse
|
46
|
Astrocytic LRP1 Mediates Brain Aβ Clearance and Impacts Amyloid Deposition. J Neurosci 2017; 37:4023-4031. [PMID: 28275161 DOI: 10.1523/jneurosci.3442-16.2017] [Citation(s) in RCA: 166] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 02/27/2017] [Accepted: 03/02/2017] [Indexed: 11/21/2022] Open
Abstract
Accumulation and deposition of amyloid-β (Aβ) in the brain represent an early and perhaps necessary step in the pathogenesis of Alzheimer's disease (AD). Aβ accumulation leads to the formation of Aβ aggregates, which may directly and indirectly lead to eventual neurodegeneration. While Aβ production is accelerated in many familial forms of early-onset AD, increasing evidence indicates that impaired clearance of Aβ is more evident in late-onset AD. To uncover the mechanisms underlying impaired Aβ clearance in AD, we examined the role of low-density lipoprotein receptor-related protein 1 (LRP1) in astrocytes. Although LRP1 has been shown to play critical roles in brain Aβ metabolism in neurons and vascular mural cells, its role in astrocytes, the most abundant cell type in the brain responsible for maintaining neuronal homeostasis, remains unclear. Here, we show that astrocytic LRP1 plays a critical role in brain Aβ clearance. LRP1 knockdown in primary astrocytes resulted in decreased cellular Aβ uptake and degradation. In addition, silencing of LRP1 in astrocytes led to downregulation of several major Aβ-degrading enzymes, including matrix metalloproteases MMP2, MMP9, and insulin-degrading enzyme. More important, conditional knock-out of the Lrp1 gene in astrocytes in the background of APP/PS1 mice impaired brain Aβ clearance, exacerbated Aβ accumulation, and accelerated amyloid plaque deposition without affecting its production. Together, our results demonstrate that astrocytic LRP1 plays an important role in Aβ metabolism and that restoring LRP1 expression and function in the brain could be an effective strategy to facilitate Aβ clearance and counter amyloid pathology in AD.SIGNIFICANCE STATEMENT Astrocytes represent a major cell type regulating brain homeostasis; however, their roles in brain clearance of amyloid-β (Aβ) and underlying mechanism are not clear. In this study, we used both cellular models and conditional knock-out mouse models to address the role of a critical Aβ receptor, the low-density lipoprotein receptor-related protein 1 (LRP1) in astrocytes. We found that LRP1 in astrocytes plays a critical role in brain Aβ clearance by modulating several Aβ-degrading enzymes and cellular degradation pathways. Our results establish a critical role of astrocytic LRP1 in brain Aβ clearance and shed light on specific Aβ clearance pathways that may help to establish new targets for AD prevention and therapy.
Collapse
|
47
|
Finan GM, Realubit R, Chung S, Lütjohann D, Wang N, Cirrito JR, Karan C, Kim TW. Bioactive Compound Screen for Pharmacological Enhancers of Apolipoprotein E in Primary Human Astrocytes. Cell Chem Biol 2016; 23:1526-1538. [DOI: 10.1016/j.chembiol.2016.10.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Revised: 09/13/2016] [Accepted: 10/25/2016] [Indexed: 02/07/2023]
|
48
|
Mohammadzadeh Honarvar N, Saedisomeolia A, Abdolahi M, Shayeganrad A, Taheri Sangsari G, Hassanzadeh Rad B, Muench G. Molecular Anti-inflammatory Mechanisms of Retinoids and Carotenoids in Alzheimer's Disease: a Review of Current Evidence. J Mol Neurosci 2016; 61:289-304. [PMID: 27864661 DOI: 10.1007/s12031-016-0857-x] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 10/21/2016] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD) is considered as one of the most prevalent neurodegenerative disorders characterized by progressive loss of mental function and ability to learn. AD is a multifactorial disorder. Various hypotheses are suggested for the pathophysiology of AD including "Aβ hypothesis," "tau hypothesis," and "cholinergic hypothesis." Recently, it has been demonstrated that neuroinflammation is involved in the pathogenesis of AD. Neuroinflammation causes synaptic dysfunction and neuronal death within the brain. Excessive production of pro-inflammatory mediators induces Aβ peptide production/accumulation and hyperphosphorylated tau generating inflammatory molecules and cytokines. These inflammatory molecules disrupt blood-brain barrier integrity and increase the production of Aβ42 oligomers. Retinoids and carotenoids are potent antioxidants and anti-inflammatory agents having neuroprotective properties. They are able to prevent disease progression through several mechanisms such as suppression of Aβ peptide production/accumulation, oxidative stress, and pro-inflammatory mediator's secretion as well as improvement of cognitive performance. These observations, therefore, confirm the neuroprotective role of retinoids and carotenoids through multiple pathways. Therefore, the administration of these nutrients is considered as a promising approach to the prevention and/or treatment of AD in the future. The aim of this review is to present existing evidences regarding the beneficial effects of retinoids and carotenoids on AD's risk and outcomes, seeking the mechanism of their action.
Collapse
Affiliation(s)
- Niyaz Mohammadzadeh Honarvar
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Saedisomeolia
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran. .,Department of Pharmacology, School of Medicine, Western Sydney University, NSW, Australia. .,School of Molecular Bioscience, Charles Perkins Centre, University of Sydney, NSW, Australia.
| | - Mina Abdolahi
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Shayeganrad
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | - Gerald Muench
- Department of Pharmacology, School of Medicine, Western Sydney University, NSW, Australia
| |
Collapse
|
49
|
Zhang X, Hu J, Zhong L, Wang N, Yang L, Liu CC, Li H, Wang X, Zhou Y, Zhang Y, Xu H, Bu G, Zhuang J. Quercetin stabilizes apolipoprotein E and reduces brain Aβ levels in amyloid model mice. Neuropharmacology 2016; 108:179-92. [DOI: 10.1016/j.neuropharm.2016.04.032] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 04/18/2016] [Accepted: 04/21/2016] [Indexed: 10/21/2022]
|
50
|
Yamazaki Y, Painter MM, Bu G, Kanekiyo T. Apolipoprotein E as a Therapeutic Target in Alzheimer's Disease: A Review of Basic Research and Clinical Evidence. CNS Drugs 2016; 30:773-89. [PMID: 27328687 PMCID: PMC5526196 DOI: 10.1007/s40263-016-0361-4] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disorder that causes progressive cognitive decline. The majority of AD cases are sporadic and late-onset (>65 years old) making it the leading cause of dementia in the elderly. While both genetic and environmental factors contribute to the development of late-onset AD (LOAD), APOE polymorphism is a major genetic risk determinant for LOAD. In humans, the APOE gene has three major allelic variants: ε2, ε3, and ε4, of which APOE ε4 is the strongest genetic risk factor for LOAD, whereas APOE ε2 is protective. Mounting evidence suggests that APOE ε4 contributes to AD pathogenesis through multiple pathways including facilitated amyloid-β deposition, increased tangle formation, synaptic dysfunction, exacerbated neuroinflammation, and cerebrovascular defects. Since APOE modulates multiple biological processes through its corresponding protein apolipoprotein E (apoE), APOE gene and apoE properties have been a promising target for therapy and drug development against AD. In this review, we summarize the current evidence regarding how the APOE ε4 allele contributes to the pathogenesis of AD and how relevant therapeutic approaches can be developed to target apoE-mediated pathways in AD.
Collapse
Affiliation(s)
- Yu Yamazaki
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Meghan M Painter
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA
| | - Takahisa Kanekiyo
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL, 32224, USA.
| |
Collapse
|