1
|
Azarfar K, Decourt B, Camacho BS, Lawrence JJ, Omondi TR, Sabbagh MN. Cholesterol-modifying strategies for Alzheimer disease: promise or fallacy? Expert Rev Neurother 2025; 25:521-535. [PMID: 40140971 PMCID: PMC12068190 DOI: 10.1080/14737175.2025.2483928] [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: 09/21/2024] [Revised: 03/06/2025] [Accepted: 03/20/2025] [Indexed: 03/28/2025]
Abstract
INTRODUCTION As the world population ages, Alzheimer disease (AD) prevalence increases. However, understanding of AD etiology continues to evolve, and the pathophysiological processes involved are only partially elucidated. One compound suspected to play a role in the development and progression of AD is cholesterol. Several lines of evidence support this connection, yet it remains unclear whether cholesterol-modifying strategies have potential applications in the clinical management of AD. AREAS COVERED A deep literature search using PubMed was performed to prepare this narrative review. The literature search, performed in early 2024, was inclusive of literature from 1990 to 2024. After providing an overview of cholesterol metabolism, this study summarizes key preclinical studies that have investigated cholesterol-modifying therapies in laboratory models of AD. It also summarizes past and current clinical trials testing specific targets modulated by anti-cholesterol therapies in AD patients. EXPERT OPINION Based on current epidemiological and mechanistic studies, cholesterol likely plays a role in AD etiology. The use of cholesterol-modifying therapies could be a promising treatment approach if administered at presymptomatic to early AD phases, but it is unlikely to be efficient in mild, moderate, and late AD stages. Several recommendations are provided for hypercholesterolemia management in AD patients.
Collapse
Affiliation(s)
- Katia Azarfar
- Department of Pharmacology and Neurosciences, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Boris Decourt
- Department of Pharmacology and Neurosciences, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Brandon Sanchez Camacho
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, Arizona
| | - John Joshua Lawrence
- Department of Pharmacology and Neurosciences, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Tania R. Omondi
- Department of Pharmacology and Neurosciences, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Marwan N. Sabbagh
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, Arizona
| |
Collapse
|
2
|
Gu RF, Hronowski X, Shao Z, Gao B, Soucey K, Sun F, Tsai HH, Wei R. Dynamic Proteome Changes in Cuprizone-Induced Demyelination and Remyelination in the Mouse Brain. J Proteome Res 2025. [PMID: 40305778 DOI: 10.1021/acs.jproteome.4c01036] [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: 05/02/2025]
Abstract
This study aimed to gain insights into the dynamic proteome changes and underlying molecular mechanisms of de/remyelination in a cuprizone model, a widely used preclinical model of multiple sclerosis (MS). Longitudinal sampling of control or cuprizone-treated mouse brains was executed at 6 time points over 6 weeks. Data analysis included 8489 quantified proteins. Differential proteomic and GO analyses revealed that 5.9% of the quantified proteome was altered, including reported and novel de/remyelination-relevant protein changes and underlying pathways. We found that oligodendrocyte proteins (Fa2h and Ugt8) were significantly changed during demyelination, suggesting that dysregulated sphingolipid metabolism in MS may stem from oligodendrocyte pathology. Importantly, we showed that the cholesterol biosynthesis pathway was the most enriched biological process in a subset of significantly changed proteins, where myelination was highly enriched. We further validated the changes in the cholesterol biosynthesis pathway through targeted GC-MS analysis of intermediate sterols, supporting the critical role of cholesterol biosynthesis in de/remyelination. Unexpectedly, changes of myelin-associated proteins, Mbp and Plp1, were minimal, while Ermn showed significant reduction tracking with demyelination, indicating that some myelin protein changes are more sensitive to demyelination. Together with a list of significantly altered proteins, the results of this study could benefit future remyelination research.
Collapse
Affiliation(s)
- Rong-Fang Gu
- Chemical Biology and Proteomics, Biogen, Cambridge, Massachusetts 02142, United States
| | - Xiaoping Hronowski
- Chemical Biology and Proteomics, Biogen, Cambridge, Massachusetts 02142, United States
| | - Zhaohui Shao
- Multiple Sclerosis Immunology Research, Biogen, Cambridge, Massachusetts 02142, United States
| | - Benbo Gao
- Chemical Biology and Proteomics, Biogen, Cambridge, Massachusetts 02142, United States
| | - Kayla Soucey
- Multiple Sclerosis Immunology Research, Biogen, Cambridge, Massachusetts 02142, United States
| | - Fangxu Sun
- Chemical Biology and Proteomics, Biogen, Cambridge, Massachusetts 02142, United States
| | - Hui-Hsin Tsai
- Multiple Sclerosis Clinical Development, Biogen, Cambridge, Massachusetts 02142, United States
| | - Ru Wei
- Chemical Biology and Proteomics, Biogen, Cambridge, Massachusetts 02142, United States
| |
Collapse
|
3
|
Durairaj P, Liu ZL. Brain Cytochrome P450: Navigating Neurological Health and Metabolic Regulation. J Xenobiot 2025; 15:44. [PMID: 40126262 PMCID: PMC11932283 DOI: 10.3390/jox15020044] [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: 01/07/2025] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 03/25/2025] Open
Abstract
Human cytochrome P450 (CYP) enzymes in the brain represent a crucial frontier in neuroscience, with far-reaching implications for drug detoxification, cellular metabolism, and the progression of neurodegenerative diseases. The brain's complex architecture, composed of interconnected cell types and receptors, drives unique neuronal signaling pathways, modulates enzyme functions, and leads to distinct CYP gene expression and regulation patterns compared to the liver. Despite their relatively low levels of expression, brain CYPs exert significant influence on drug responses, neurotoxin susceptibility, behavior, and neurological disease risk. These enzymes are essential for maintaining brain homeostasis, mediating cholesterol turnover, and synthesizing and metabolizing neurochemicals, neurosteroids, and neurotransmitters. Moreover, they are key participants in oxidative stress responses, neuroprotection, and the regulation of inflammation. In addition to their roles in metabolizing psychotropic drugs, substances of abuse, and endogenous compounds, brain CYPs impact drug efficacy, safety, and resistance, underscoring their importance beyond traditional drug metabolism. Their involvement in critical physiological processes also links them to neuroprotection, with significant implications for the onset and progression of neurodegenerative diseases. Understanding the roles of cerebral CYP enzymes is vital for advancing neuroprotective strategies, personalizing treatments for brain disorders, and developing CNS-targeting therapeutics. This review explores the emerging roles of CYP enzymes, particularly those within the CYP1-3 and CYP46 families, highlighting their functional diversity and the pathological consequences of their dysregulation on neurological health. It also examines the potential of cerebral CYP-based biomarkers to improve the diagnosis and treatment of neurodegenerative disorders, offering new avenues for therapeutic innovation.
Collapse
Affiliation(s)
- Pradeepraj Durairaj
- Department of Chemical and Biomedical Engineering, Florida State University, Tallahassee, FL 32310, USA
- Department of Chemical and Biomedical Engineering, Florida A&M University, Tallahassee, FL 32310, USA
| | - Zixiang Leonardo Liu
- Department of Chemical and Biomedical Engineering, Florida State University, Tallahassee, FL 32310, USA
- Department of Chemical and Biomedical Engineering, Florida A&M University, Tallahassee, FL 32310, USA
- Institute for Successful Longevity, Florida State University, Tallahassee, FL 32310, USA
| |
Collapse
|
4
|
Wheless JW, Rho JM. Role of cholesterol in modulating brain hyperexcitability. Epilepsia 2025; 66:33-46. [PMID: 39487852 PMCID: PMC11742637 DOI: 10.1111/epi.18174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 10/15/2024] [Accepted: 10/15/2024] [Indexed: 11/04/2024]
Abstract
Cholesterol is a critical molecule in the central nervous system, and imbalances in the synthesis and metabolism of brain cholesterol can result in a range of pathologies, including those related to hyperexcitability. The impact of cholesterol on disorders of epilepsy and developmental and epileptic encephalopathies is an area of growing interest. Cholesterol cannot cross the blood-brain barrier, and thus the brain synthesizes and metabolizes its own pool of cholesterol. The primary metabolic enzyme for brain cholesterol is cholesterol 24-hydroxylase (CH24H), which metabolizes cholesterol into 24S-hydroxycholesterol (24HC). Dysregulation of CH24H and 24HC can affect neuronal excitability through a range of mechanisms. 24HC is a positive allosteric modulator of N-methyl-D-aspartate (NMDA) receptors and can increase glutamate release via tumor necrosis factor-α-dependent pathways. Increasing cholesterol metabolism can lead to dysfunction of excitatory amino acid transporter 2 and impair glutamate reuptake. Finally, overstimulation of NMDA receptors can further activate metabolism of cholesterol, leading to a vicious cycle of overactivation. All of these mechanisms increase extracellular glutamate and can lead to hyperexcitability. For these reasons, the cholesterol pathway represents a new potential mechanistic target for antiseizure medications. CH24H inhibition has been shown to decrease seizure behavior and improve survival in multiple animal models of epilepsy and could be a promising new mechanism of action for the treatment of neuronal hyperexcitability and developmental and epileptic encephalopathies.
Collapse
Affiliation(s)
- James W. Wheless
- Division of Pediatric NeurologyUniversity of Tennessee Health Science CenterMemphisTennesseeUSA
| | - Jong M. Rho
- Department of Neurosciences, Pediatrics and PharmacologyUniversity of California San Diego School of MedicineSan DiegoCaliforniaUSA
- Rady Children's Hospital–San DiegoSan DiegoCaliforniaUSA
| |
Collapse
|
5
|
Boyarko B, Podvin S, Greenberg B, Arnold S, Juanes AM, van der Kant R, Goldstein L, Momper JD, Bang A, Silverman J, Feldman HH, Hook V. Challenges and Opportunities for Consideration of Efavirenz Drug Repurposing for Alzheimer's Disease Therapeutics. ACS Pharmacol Transl Sci 2024; 7:2924-2935. [PMID: 39421657 PMCID: PMC11480897 DOI: 10.1021/acsptsci.4c00229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 08/01/2024] [Accepted: 08/05/2024] [Indexed: 10/19/2024]
Abstract
Therapeutic research and development for Alzheimer's disease (AD) has been an area of intense research to alleviate memory loss and neurodegeneration. There is growing interest in drug repositioning and repurposing strategies for FDA-approved medications as potential candidates that may further advance AD therapeutics. The FDA drug efavirenz has been investigated as a candidate drug for repurposing as an AD medication. The proposed mechanism of action of efavirenz (at low doses) is the activation of the neuron-specific enzyme CYP46A1 that converts excess brain cholesterol into 24-hydroxycholesterol (24-HC) that is exported to the periphery. Efavirenz at a low dose was found to improve memory deficit in the 5XFAD model of AD that was accompanied by elevated 24-HC and reduction in Aβ; furthermore, efavirenz reduced pTau and excess cholesterol levels in human iPSC-derived Alzheimer's neurons. The low dose of efavirenz used in the AD mouse model to increase 24-HC contrasts with the use of more than 100-fold higher doses of efavirenz for clinical treatment of human immunodeficiency virus (HIV) through inhibition of reverse transcriptase. Low doses of efavirenz may avoid neurotoxic adverse effects that occur at high efavirenz doses used for HIV treatment. This review evaluates the drug properties of efavirenz with respect to its preclinical data on regulating memory deficit, pharmacokinetics, pharmacodynamics, metabolites, and genetic variabilities in drug metabolism as well as its potential adverse effects. These analyses discuss the challenges and questions that should be addressed in future studies to consider the opportunity for low dose efavirenz as a candidate for AD drug development.
Collapse
Affiliation(s)
- Ben Boyarko
- Skaggs
School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92093, United States
- Alzheimer’s
Disease Cooperative Study, School of Medicine, University of California, San Diego, La Jolla, California 92093, United States
| | - Sonia Podvin
- Skaggs
School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92093, United States
| | - Barry Greenberg
- Department
of Neurology, Johns Hopkins University School
of Medicine, Baltimore, Maryland 21287, United States
| | - Steven Arnold
- Alzheimer’s
Clinical and Translational Research Unit, Massachusetts General Hospital, Charlestown, Massachusetts 02129, United States
| | - Almudena Maroto Juanes
- Department
of Functional Genomics, Center for Neurogenomics and Cognitive Research,
Amsterdam Neuroscience, VU University Amsterdam
de Boelelaan, Amsterdam 1081 HV, The Netherlands
| | - Rik van der Kant
- Department
of Functional Genomics, Center for Neurogenomics and Cognitive Research,
Amsterdam Neuroscience, VU University Amsterdam
de Boelelaan, Amsterdam 1081 HV, The Netherlands
| | - Lawrence Goldstein
- Department
of Cellular and Molecular Medicine, University
of California, San Diego, La Jolla, California 92093, United States
| | - Jeremiah D. Momper
- Skaggs
School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92093, United States
| | - Anne Bang
- Conrad
Prebys Center for Chemical Genomics, Sanford
Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, United States
| | - James Silverman
- Alzheimer’s
Disease Cooperative Study, School of Medicine, University of California, San Diego, La Jolla, California 92093, United States
- Department
of Neurosciences, University of California,
San Diego, La Jolla, California 92093, United States
| | - Howard H. Feldman
- Alzheimer’s
Disease Cooperative Study, School of Medicine, University of California, San Diego, La Jolla, California 92093, United States
- Department
of Neurosciences, University of California,
San Diego, La Jolla, California 92093, United States
| | - Vivian Hook
- Skaggs
School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California 92093, United States
- Alzheimer’s
Disease Cooperative Study, School of Medicine, University of California, San Diego, La Jolla, California 92093, United States
- Department
of Neurosciences, University of California,
San Diego, La Jolla, California 92093, United States
| |
Collapse
|
6
|
Mast N, Butts M, Pikuleva IA. Unbiased insights into the multiplicity of the CYP46A1 brain effects in 5XFAD mice treated with low dose-efavirenz. J Lipid Res 2024; 65:100555. [PMID: 38719151 PMCID: PMC11176809 DOI: 10.1016/j.jlr.2024.100555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 03/12/2024] [Accepted: 05/01/2024] [Indexed: 05/30/2024] Open
Abstract
Cytochrome P450 46A1 (CYP46A1) is the CNS-specific cholesterol 24-hydroxylase that controls cholesterol elimination and turnover in the brain. In mouse models, pharmacologic CYP46A1 activation with low-dose efavirenz or by gene therapy mitigates the manifestations of various brain disorders, neurologic, and nonneurologic, by affecting numerous, apparently unlinked biological processes. Accordingly, CYP46A1 is emerging as a promising therapeutic target; however, the mechanisms underlying the multiplicity of the brain CYP46A1 activity effects are currently not understood. We proposed the chain reaction hypothesis, according to which CYP46A1 is important for the three primary (unifying) processes in the brain (sterol flux through the plasma membranes, acetyl-CoA, and isoprenoid production), which in turn affect a variety of secondary processes. We already identified several processes secondary to changes in sterol flux and herein undertook a multiomics approach to compare the brain proteome, acetylproteome, and metabolome of 5XFAD mice (an Alzheimer's disease model), control and treated with low-dose efavirenz. We found that the latter had increased production of phospholipids from the corresponding lysophospholipids and a globally increased protein acetylation (including histone acetylation). Apparently, these effects were secondary to increased acetyl-CoA production. Signaling of small GTPases due to their altered abundance or abundance of their regulators could be affected as well, potentially via isoprenoid biosynthesis. In addition, the omics data related differentially abundant molecules to other biological processes either reported previously or new. Thus, we obtained unbiased mechanistic insights and identified potential players mediating the multiplicity of the CYP46A1 brain effects and further detailed our chain reaction hypothesis.
Collapse
Affiliation(s)
- Natalia Mast
- Department of Ophthalmology and Visual Science, Case Western Reserve University, Cleveland, OH, USA
| | - Makaya Butts
- Department of Ophthalmology and Visual Science, Case Western Reserve University, Cleveland, OH, USA
| | - Irina A Pikuleva
- Department of Ophthalmology and Visual Science, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
7
|
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
|
8
|
Shu HJ, Ziolkowski LH, Salvatore SV, Benz AM, Wozniak DF, Yuede CM, Paul SM, Zorumski CF, Mennerick S. Effects of Complete and Partial Loss of the 24S-Hydroxycholesterol-Generating Enzyme Cyp46a1 on Behavior and Hippocampal Transcription in Mouse. Biomolecules 2024; 14:254. [PMID: 38540675 PMCID: PMC10968171 DOI: 10.3390/biom14030254] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/09/2024] [Accepted: 02/15/2024] [Indexed: 12/09/2024] Open
Abstract
Brain cholesterol metabolic products include neurosteroids and oxysterols, which play important roles in cellular physiology. In neurons, the cholesterol oxidation product, 24S-hydroxycholesterol (24S-HC), is a regulator of signaling and transcription. Here, we examined the behavioral effects of 24S-HC loss, using global and cell-selective genetic deletion of the synthetic enzyme CYP46A1. Mice that are globally deficient in CYP46A1 exhibited hypoactivity at young ages and unexpected increases in conditioned fear memory. Despite strong reductions in hippocampal 24S-HC in mice with selective loss of CYP46A1 in VGLUT1-positive cells, behavioral effects were not recapitulated in these conditional knockout mice. Global knockout produced strong, developmentally dependent transcriptional effects on select cholesterol metabolism genes. These included paradoxical changes in Liver X Receptor targets. Again, conditional knockout was insufficient to recapitulate most changes. Overall, our results highlight the complex effects of 24S-HC in an in vivo setting that are not fully predicted by known mechanisms. The results also demonstrate that the complete inhibition of enzymatic activity may be needed for a detectable, therapeutically relevant impact on gene expression and behavior.
Collapse
Affiliation(s)
- Hong-Jin Shu
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA (S.V.S.); (D.F.W.); (C.M.Y.); (S.M.P.)
| | - Luke H. Ziolkowski
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA (S.V.S.); (D.F.W.); (C.M.Y.); (S.M.P.)
| | - Sofia V. Salvatore
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA (S.V.S.); (D.F.W.); (C.M.Y.); (S.M.P.)
| | - Ann M. Benz
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA (S.V.S.); (D.F.W.); (C.M.Y.); (S.M.P.)
| | - David F. Wozniak
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA (S.V.S.); (D.F.W.); (C.M.Y.); (S.M.P.)
- Taylor Family Institute for Innovative Psychiatry Research, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Carla M. Yuede
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA (S.V.S.); (D.F.W.); (C.M.Y.); (S.M.P.)
| | - Steven M. Paul
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA (S.V.S.); (D.F.W.); (C.M.Y.); (S.M.P.)
- Taylor Family Institute for Innovative Psychiatry Research, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Charles F. Zorumski
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA (S.V.S.); (D.F.W.); (C.M.Y.); (S.M.P.)
- Taylor Family Institute for Innovative Psychiatry Research, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Steven Mennerick
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA (S.V.S.); (D.F.W.); (C.M.Y.); (S.M.P.)
- Taylor Family Institute for Innovative Psychiatry Research, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
9
|
Tripodi D, Vitarelli F, Spiti S, Leoni V. The Diagnostic Use of the Plasma Quantification of 24S-Hydroxycholesterol and Other Oxysterols in Neurodegenerative Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1440:337-351. [PMID: 38036888 DOI: 10.1007/978-3-031-43883-7_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
Cholesterol regulates fluidity and structure of cellular membranes. The brain is involved in signal transduction, synaptogenesis, and membrane trafficking. An impairment of its metabolism was observed in different neurodegenerative diseases, such as Multiple Sclerosis, Alzheimer, and Huntington diseases. Because of the blood-brain barrier, cholesterol cannot be uptaken from the circulation and all the cholesterol is locally synthetized. The excess cholesterol in neurons is converted into 24S-hydroxycholesterol (24OHC) by the cholesterol 24-hydroxylase (CYP46A1). The plasmatic concentration of 24OHC results in the balance between cerebral production and liver elimination. It is related to the number of metabolically active neurons in the brain. Several factors that affect the brain cholesterol turnover and the liver elimination of oxysterols, the genetic background, nutrition, and lifestyle habits were found to significantly affect plasma levels of 24OHC. Reduced levels of 24OHC were found related to the loss of metabolically active cells and the degree of brain atrophy. The dysfunction of the blood-brain barrier, inflammation, and increased cholesterol turnover might overlap with this progressive reduction giving temporary increased levels of 24OHC.The study of plasma 24OHC is likely to offer an insight into brain cholesterol turnover with a limited diagnostic power.
Collapse
Affiliation(s)
- Domenico Tripodi
- Laboratory of Clinical Pathology, Hospital Pio XI of Desio, ASST-Brianza and Department of Medicine and Surgery, University of Milano Bicocca, Desio, MB, Italy
| | - Federica Vitarelli
- Laboratory of Clinical Pathology, Hospital Pio XI of Desio, ASST-Brianza and Department of Medicine and Surgery, University of Milano Bicocca, Desio, MB, Italy
| | - Simona Spiti
- Laboratory of Clinical Pathology, Hospital Pio XI of Desio, ASST-Brianza and Department of Medicine and Surgery, University of Milano Bicocca, Desio, MB, Italy
| | - Valerio Leoni
- Laboratory of Clinical Pathology, Hospital Pio XI of Desio, ASST-Brianza and Department of Medicine and Surgery, University of Milano Bicocca, Desio, MB, Italy.
| |
Collapse
|
10
|
Paseban T, Alavi MS, Etemad L, Roohbakhsh A. The role of the ATP-Binding Cassette A1 (ABCA1) in neurological disorders: a mechanistic review. Expert Opin Ther Targets 2023; 27:531-552. [PMID: 37428709 DOI: 10.1080/14728222.2023.2235718] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 07/09/2023] [Indexed: 07/12/2023]
Abstract
INTRODUCTION Cholesterol homeostasis is critical for normal brain function. It is tightly controlled by various biological elements. ATP-binding cassette transporter A1 (ABCA1) is a membrane transporter that effluxes cholesterol from cells, particularly astrocytes, into the extracellular space. The recent studies pertaining to ABCA1's role in CNS disorders were included in this study. AREAS COVERED In this comprehensive literature review, preclinical and human studies showed that ABCA1 has a significant role in the following diseases or disorders: Alzheimer's disease, Parkinson's disease, Huntington's disease, multiple sclerosis, neuropathy, anxiety, depression, psychosis, epilepsy, stroke, and brain ischemia and trauma. EXPERT OPINION ABCA1 via modulating normal and aberrant brain functions such as apoptosis, phagocytosis, BBB leakage, neuroinflammation, amyloid β efflux, myelination, synaptogenesis, neurite outgrowth, and neurotransmission promotes beneficial effects in aforementioned diseases. ABCA1 is a key molecule in the CNS. By boosting its expression or function, some CNS disorders may be resolved. In preclinical studies, liver X receptor agonists have shown promise in treating CNS disorders via ABCA1 and apoE enhancement.
Collapse
Affiliation(s)
- Tahere Paseban
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohaddeseh Sadat Alavi
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Leila Etemad
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Roohbakhsh
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|