1
|
Preethy H A, Rajendran K, Sukumar AJ, Krishnan UM. Emerging paradigms in Alzheimer's therapy. Eur J Pharmacol 2024; 981:176872. [PMID: 39117266 DOI: 10.1016/j.ejphar.2024.176872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 07/13/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024]
Abstract
Alzheimer's disease is a neurodegenerative disorder that affects elderly, and its incidence is continuously increasing across the globe. Unfortunately, despite decades of research, a complete cure for Alzheimer's disease continues to elude us. The current medications are mainly symptomatic and slow the disease progression but do not result in reversal of all disease pathologies. The growing body of knowledge on the factors responsible for the onset and progression of the disease has resulted in the identification of new targets that could be targeted for treatment of Alzheimer's disease. This has opened new vistas for treatment of Alzheimer's disease that have moved away from chemotherapeutic agents modulating a single target to biologics and combinations that acted on multiple targets thereby offering better therapeutic outcomes. This review discusses the emerging directions in therapeutic interventions against Alzheimer's disease highlighting their merits that promise to change the treatment paradigm and challenges that limit their clinical translation.
Collapse
Affiliation(s)
- Agnes Preethy H
- School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, India; Centre for Nanotechnology & Advanced Biomaterials, SASTRA Deemed University, Thanjavur, India
| | - Kayalvizhi Rajendran
- School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, India; Centre for Nanotechnology & Advanced Biomaterials, SASTRA Deemed University, Thanjavur, India
| | - Anitha Josephine Sukumar
- School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, India; Centre for Nanotechnology & Advanced Biomaterials, SASTRA Deemed University, Thanjavur, India
| | - Uma Maheswari Krishnan
- School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, India; Centre for Nanotechnology & Advanced Biomaterials, SASTRA Deemed University, Thanjavur, India; School of Arts, Sciences, Humanities & Education, SASTRA Deemed University, Thanjavur, India.
| |
Collapse
|
2
|
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
|
3
|
Ahmed H, Wang Y, Griffiths WJ, Levey AI, Pikuleva I, Liang SH, Haider A. Brain cholesterol and Alzheimer's disease: challenges and opportunities in probe and drug development. Brain 2024; 147:1622-1635. [PMID: 38301270 PMCID: PMC11068113 DOI: 10.1093/brain/awae028] [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: 03/04/2023] [Revised: 12/20/2023] [Accepted: 01/13/2024] [Indexed: 02/03/2024] Open
Abstract
Cholesterol homeostasis is impaired in Alzheimer's disease; however, attempts to modulate brain cholesterol biology have not translated into tangible clinical benefits for patients to date. Several recent milestone developments have substantially improved our understanding of how excess neuronal cholesterol contributes to the pathophysiology of Alzheimer's disease. Indeed, neuronal cholesterol was linked to the formation of amyloid-β and neurofibrillary tangles through molecular pathways that were recently delineated in mechanistic studies. Furthermore, remarkable advances in translational molecular imaging have now made it possible to probe cholesterol metabolism in the living human brain with PET, which is an important prerequisite for future clinical trials that target the brain cholesterol machinery in Alzheimer's disease patients-with the ultimate aim being to develop disease-modifying treatments. This work summarizes current concepts of how the biosynthesis, transport and clearance of brain cholesterol are affected in Alzheimer's disease. Further, current strategies to reverse these alterations by pharmacotherapy are critically discussed in the wake of emerging translational research tools that support the assessment of brain cholesterol biology not only in animal models but also in patients with Alzheimer's disease.
Collapse
Affiliation(s)
- Hazem Ahmed
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, Yale University, New Haven, CT 06510, USA
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Institute of Pharmaceutical Sciences ETH, 8093 Zurich, Switzerland
| | - Yuqin Wang
- Institute of Life Science, Swansea University Medical School, Swansea SA2 8PP, UK
| | - William J Griffiths
- Institute of Life Science, Swansea University Medical School, Swansea SA2 8PP, UK
| | - Allan I Levey
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Irina Pikuleva
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Steven H Liang
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Ahmed Haider
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| |
Collapse
|
4
|
Feng Y, Gong C, Zhu J, Liu G, Tang Y, Li W. Unraveling the Ligand-Binding Sites of CYP3A4 by Molecular Dynamics Simulations with Solvent Probes. J Chem Inf Model 2024; 64:3451-3464. [PMID: 38593186 DOI: 10.1021/acs.jcim.4c00089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
Cytochrome P450 3A4 (CYP3A4) is one of the most important drug-metabolizing enzymes in the human body and is well known for its complicated, atypical kinetic characteristics. The existence of multiple ligand-binding sites in CYP3A4 has been widely recognized as being capable of interfering with the active pocket through allosteric effects. The identification of ligand-binding sites other than the canonical active site above the heme is especially important for understanding the atypical kinetic characteristics of CYP3A4 and the intriguing association between the ligand and the receptor. In this study, we first employed mixed-solvent molecular dynamics (MixMD) simulations coupled with the online computational predictive tools to explore potential ligand-binding sites in CYP3A4. The MixMD approach demonstrates better performance in dealing with the receptor flexibility compared with other computational tools. From the sites identified by MixMD, we then picked out multiple sites for further exploration using ensemble docking and conventional molecular dynamics (cMD) simulations. Our results indicate that three extra sites are suitable for ligand binding in CYP3A4, including one experimentally confirmed site and two novel sites.
Collapse
Affiliation(s)
- Yanjun Feng
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Changda Gong
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Jieyu Zhu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Guixia Liu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Yun Tang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Weihua Li
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| |
Collapse
|
5
|
Mast N, Li Y, Pikuleva IA. 7,8-Dihydroxy Efavirenz Is Not as Effective in CYP46A1 Activation In Vivo as Efavirenz or Its 8,14-Dihydroxy Metabolite. Int J Mol Sci 2024; 25:2242. [PMID: 38396919 PMCID: PMC10889178 DOI: 10.3390/ijms25042242] [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: 12/19/2023] [Revised: 02/08/2024] [Accepted: 02/10/2024] [Indexed: 02/25/2024] Open
Abstract
High dose (S)-efavirenz (EFV) inhibits the HIV reverse transcriptase enzyme and is used to lower HIV load. Low-dose EFV allosterically activates CYP46A1, the key enzyme for cholesterol elimination from the brain, and is investigated as a potential treatment for Alzheimer's disease. Simultaneously, we evaluate EFV dihydroxymetabolites for in vivo brain effects to compare with those of (S)-EFV. We have already tested (rac)-8,14dihydroxy EFV on 5XFAD mice, a model of Alzheimer's disease. Herein, we treated 5XFAD mice with (rac)-7,8dihydroxy EFV. In both sexes, the treatment modestly activated CYP46A1 in the brain and increased brain content of acetyl-CoA and acetylcholine. Male mice also showed a decrease in the brain levels of insoluble amyloid β40 peptides. However, the treatment had no effect on animal performance in different memory tasks. Thus, the overall brain effects of (rac)-7,8dihydroxy EFV were weaker than those of EFV and (rac)-8,14dihydroxy EFV and did not lead to cognitive improvements as were seen in treatments with EFV and (rac)-8,14dihydroxy EFV. An in vitro study assessing CYP46A1 activation in co-incubations with EFV and (rac)-7,8dihydroxy EFV or (rac)-8,14dihydroxy EFV was carried out and provided insight into the compound doses and ratios that could be used for in vivo co-treatments with EFV and its dihydroxymetabolite.
Collapse
Affiliation(s)
| | | | - Irina A. Pikuleva
- Department of Ophthalmology and Visual Science, Case Western Reserve University, Cleveland, OH 44106, USA; (N.M.); (Y.L.)
| |
Collapse
|
6
|
Lam M, Kuo SY, Reis S, Gestwicki JE, Silva MC, Haggarty SJ. Cholesterol Dysregulation Drives Seed-Dependent Tau Aggregation in Patient Stem Cell-Derived Models of Tauopathy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.11.571147. [PMID: 38168389 PMCID: PMC10759997 DOI: 10.1101/2023.12.11.571147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Tauopathies are a class of neurodegenerative diseases characterized by the progressive misfolding and accumulation of pathological tau protein in focal regions of the brain, leading to insidious neurodegeneration. Abnormalities in cholesterol metabolism and homeostasis have also been implicated in various neurodegenerative diseases. However, the connection between cholesterol dysregulation and tau pathology remains largely unknown. To model and measure the impact of cholesterol dysregulation on tau, we utilized a combination of in vitro and ex vivo tau aggregation assays using an engineered tau biosensor cell line and human induced pluripotent stem cell (iPSC)-derived neuronal cultures from an individual harboring an autosomal dominant P301L tau mutation and from a healthy control. We demonstrate that excess cholesterol esters lead to an increased rate of tau aggregation in vitro and an increase in seed-dependent insoluble tau aggregates detected in the biosensor line. We observed a strong correlation between cholesterol ester concentration and the presence of high-molecular-weight, oligomeric tau species. Importantly, in tauopathy patient iPSC-derived neurons harboring a P301L tau mutation with endogenous forms of misfolded tau, we show that acute dysregulation of cholesterol homeostasis through acute exposure to human plasma-purified cholesterol esters formed by the linkage of fatty acids to the hydroxyl group of cholesterol leads to the rapid accumulation of phosphorylated tau. Conversely, treatment with the same cholesterol esters pool did not lead to subsequent accumulation of phosphorylated tau in control iPSC-derived neurons. Finally, treatment with a heterobifunctional, small-molecule degrader designed to selectively engage and catalyze the ubiquitination and proteasomal degradation of aberrant tau species prevented cholesterol ester-induced aggregation of tau in the biosensor cell line in a Cereblon E3 ligase-dependent manner. Degrader treatment also restored the resiliency of tauopathy patient-derived neurons towards cholesterol ester-induced tau aggregation phenotypes. Taken together, our study supports a key role of cholesterol dysregulation in tau aggregation. Moreover, it provides further pre-clinical validation of the therapeutic strategy of targeted protein degradation with heterobifunctional tau degraders for blocking tau seeding.
Collapse
|
7
|
Valenza M, Birolini G, Cattaneo E. The translational potential of cholesterol-based therapies for neurological disease. Nat Rev Neurol 2023; 19:583-598. [PMID: 37644213 DOI: 10.1038/s41582-023-00864-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2023] [Indexed: 08/31/2023]
Abstract
Cholesterol is an important metabolite and membrane component and is enriched in the brain owing to its role in neuronal maturation and function. In the adult brain, cholesterol is produced locally, predominantly by astrocytes. When cholesterol has been used, recycled and catabolized, the derivatives are excreted across the blood-brain barrier. Abnormalities in any of these steps can lead to neurological dysfunction. Here, we examine how precise interactions between cholesterol production and its use and catabolism in neurons ensures cholesterol homeostasis to support brain function. As an example of a neurological disease associated with cholesterol dyshomeostasis, we summarize evidence from animal models of Huntington disease (HD), which demonstrate a marked reduction in cholesterol biosynthesis with clinically relevant consequences for synaptic activity and cognition. In addition, we examine the relationship between cholesterol loss in the brain and cognitive decline in ageing. We then present emerging therapeutic strategies to restore cholesterol homeostasis, focusing on evidence from HD mouse models.
Collapse
Affiliation(s)
- Marta Valenza
- Department of Biosciences, University of Milan, Milan, Italy.
- Istituto Nazionale di Genetica Molecolare 'Romeo ed Enrica Invernizzi', Milan, Italy.
| | - Giulia Birolini
- Department of Biosciences, University of Milan, Milan, Italy
- Istituto Nazionale di Genetica Molecolare 'Romeo ed Enrica Invernizzi', Milan, Italy
| | - Elena Cattaneo
- Department of Biosciences, University of Milan, Milan, Italy.
- Istituto Nazionale di Genetica Molecolare 'Romeo ed Enrica Invernizzi', Milan, Italy.
| |
Collapse
|
8
|
Alavi MS, Karimi G, Ghanimi HA, Roohbakhsh A. The potential of CYP46A1 as a novel therapeutic target for neurological disorders: An updated review of mechanisms. Eur J Pharmacol 2023; 949:175726. [PMID: 37062503 DOI: 10.1016/j.ejphar.2023.175726] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 04/03/2023] [Accepted: 04/14/2023] [Indexed: 04/18/2023]
Abstract
Cholesterol is a key component of the cell membrane that impacts the permeability, fluidity, and functions of membrane-bound proteins. It also participates in synaptogenesis, synaptic function, axonal growth, dendrite outgrowth, and microtubule stability. Cholesterol biosynthesis and metabolism are in balance in the brain. Its metabolism in the brain is mediated mainly by CYP46A1 or cholesterol 24-hydroxylase. It is responsible for eliminating about 80% of the cholesterol excess from the human brain. CYP46A1 converts cholesterol to 24S-hydroxycholesterol (24HC) that readily crosses the blood-brain barrier and reaches the liver for the final elimination process. Studies show that cholesterol and 24HC levels change during neurological diseases and conditions. So, it was hypothesized that inhibition or activation of CYP46A1 would be an effective therapeutic strategy. Accordingly, preclinical studies, using genetic and pharmacological interventions, assessed the role of CYP46A1 in main neurodegenerative disorders such as Parkinson's disease, Huntington's disease, Alzheimer's disease, multiple sclerosis, spinocerebellar ataxias, and amyotrophic lateral sclerosis. In addition, its role in seizures and brain injury was evaluated. The recent development of soticlestat, as a selective and potent CYP46A1 inhibitor, with significant anti-seizure effects in preclinical and clinical studies, suggests the importance of this target for future drug developments. Previous studies have shown that both activation and inhibition of CYP46A1 are of therapeutic value. This article, using recent studies, highlights the role of CYP46A1 in various brain diseases and insults.
Collapse
Affiliation(s)
- Mohaddeseh Sadat Alavi
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gholamreza Karimi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Ali Roohbakhsh
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
9
|
Zhao T, Zhong R, Zhang X, Li G, Zhou C, Fang S, Ding Y, Lin W. Efavirenz restored NMDA receptor dysfunction and inhibited epileptic seizures in GluN2A/Grin2a mutant mice. Front Neurosci 2023; 17:1086462. [PMID: 36937661 PMCID: PMC10017539 DOI: 10.3389/fnins.2023.1086462] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 02/02/2023] [Indexed: 03/06/2023] Open
Abstract
Introduction N-methyl-D-aspartate receptor (NMDAR) is one of the main receptor of the excitatory neurotransmitter glutamate in the brain, which is the key determinant of the excitatory/inhibitory balance of neural network. GluN2A/GRIN2A is one of the subunits of NMDAR and plays an important role in epilepsy. Approximately 78% of patients with GluN2A/Grin2a mutations have epilepsy, and the underlying mechanism of this association is not well characterized. Methods We constructed a mouse model of hyperthermic seizure, and conducted in vitro and in vivo electrophysiological and behavioral studies to clarify the pathogenic characteristics and mechanism of GluN2A/GRIN2A-V685G mutation. In addition, the drug efavirenz (EFV), which is used to treat HIV infection, was administrated to mutant animals to assess whether it can restore the loss of function. Results Mutant mice showed no significant change in the mRNA or protein expressions of NMDAR compared with wild type (WT) mice. Mice with GluN2A/GRIN2A-V685G mutation exhibited shorter latency to seizure, increased frequency of seizure-like events, decreased peak current and current area of NMDAR excitatory postsynaptic current, and decreased event frequency of micro-inhibitory postsynaptic current, compared to WT mice. They also exhibited decreased threshold, increased amplitude, increased input resistance, and increased root number of action potential. EFV administration reversed these changes. The loss-of-function (LoF) mutation of NMDAR changed the excitatory/inhibitory balance of neural network, rendering animal more prone to seizures. Discussion EFV was indicated to hold its potential in the treatment of inherited epilepsy.
Collapse
Affiliation(s)
- Teng Zhao
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Rui Zhong
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Xinyue Zhang
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Guangjian Li
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Chunkui Zhou
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Shaokuan Fang
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
- Shaokuan Fang,
| | - Ying Ding
- Department of Radiology, The First Hospital of Jilin University, Changchun, China
- Ying Ding,
| | - Weihong Lin
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
- *Correspondence: Weihong Lin,
| |
Collapse
|
10
|
Identification of potential inhibitors of brain-specific CYP46A1 from phytoconstituents in Indian traditional medicinal plants. JOURNAL OF PROTEINS AND PROTEOMICS 2022; 13:227-245. [PMCID: PMC9667835 DOI: 10.1007/s42485-022-00098-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 07/27/2022] [Accepted: 09/29/2022] [Indexed: 11/17/2022]
|
11
|
Haider A, Zhao C, Wang L, Xiao Z, Rong J, Xia X, Chen Z, Pfister SK, Mast N, Yutuc E, Chen J, Li Y, Shao T, Warnock GI, Dawoud A, Connors TR, Oakley DH, Wei H, Wang J, Zheng Z, Xu H, Davenport AT, Daunais JB, Van RS, Shao Y, Wang Y, Zhang MR, Gebhard C, Pikuleva I, Levey AI, Griffiths WJ, Liang SH. Assessment of cholesterol homeostasis in the living human brain. Sci Transl Med 2022; 14:eadc9967. [PMID: 36197966 PMCID: PMC9581941 DOI: 10.1126/scitranslmed.adc9967] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Alterations in brain cholesterol homeostasis have been broadly implicated in neurological disorders. Notwithstanding the complexity by which cholesterol biology is governed in the mammalian brain, excess neuronal cholesterol is primarily eliminated by metabolic clearance via cytochrome P450 46A1 (CYP46A1). No methods are currently available for visualizing cholesterol metabolism in the living human brain; therefore, a noninvasive technology that quantitatively measures the extent of brain cholesterol metabolism via CYP46A1 could broadly affect disease diagnosis and treatment options using targeted therapies. Here, we describe the development and testing of a CYP46A1-targeted positron emission tomography (PET) tracer, 18F-CHL-2205 (18F-Cholestify). Our data show that PET imaging readouts correlate with CYP46A1 protein expression and with the extent to which cholesterol is metabolized in the brain, as assessed by cross-species postmortem analyses of specimens from rodents, nonhuman primates, and humans. Proof of concept of in vivo efficacy is provided in the well-established 3xTg-AD murine model of Alzheimer's disease (AD), where we show that the probe is sensitive to differences in brain cholesterol metabolism between 3xTg-AD mice and control animals. Furthermore, our clinical observations point toward a considerably higher baseline brain cholesterol clearance via CYP46A1 in women, as compared to age-matched men. These findings illustrate the vast potential of assessing brain cholesterol metabolism using PET and establish PET as a sensitive tool for noninvasive assessment of brain cholesterol homeostasis in the clinic.
Collapse
Affiliation(s)
- Ahmed Haider
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA
- Emory University, Department of Radiology and Imaging Sciences, 1364 Clifton Rd, Atlanta, GA 30322, USA
| | - Chunyu Zhao
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA
- Emory University, Department of Radiology and Imaging Sciences, 1364 Clifton Rd, Atlanta, GA 30322, USA
| | - Lu Wang
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Zhiwei Xiao
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA
- Emory University, Department of Radiology and Imaging Sciences, 1364 Clifton Rd, Atlanta, GA 30322, USA
| | - Jian Rong
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA
- Emory University, Department of Radiology and Imaging Sciences, 1364 Clifton Rd, Atlanta, GA 30322, USA
| | - Xiaotian Xia
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, China
| | - Zhen Chen
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA
| | - Stefanie K. Pfister
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA
| | - Natalia Mast
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Eylan Yutuc
- Institute of Life Science, Swansea University Medical School, SA2 8PP Swansea, Wales, United Kingdom
| | - Jiahui Chen
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA
- Emory University, Department of Radiology and Imaging Sciences, 1364 Clifton Rd, Atlanta, GA 30322, USA
| | - Yinlong Li
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA
- Emory University, Department of Radiology and Imaging Sciences, 1364 Clifton Rd, Atlanta, GA 30322, USA
| | - Tuo Shao
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA
| | - Geoffrey I. Warnock
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Alyaa Dawoud
- Biochemistry Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, 11835, Cairo, Egypt
| | - Theresa R. Connors
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Massachusetts Alzheimer’s Disease Research Center, Boston, MA 02129, USA
| | - Derek H. Oakley
- Harvard Medical School, Boston, Massachusetts 02115, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114-2696, USA
- C.S. Kubik Laboratory for Neuropathology, Massachusetts General Hospital, Boston, MA 02114, USA
- Massachusetts Alzheimer’s Disease Research Center, Charlestown, MA 02129, USA
| | - Huiyi Wei
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Jinghao Wang
- Department of Pharmacy, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Zhihua Zheng
- Guangdong Province Pharmaceutical Association, Guangzhou 510080, China
| | - Hao Xu
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - April T. Davenport
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston Salem, NC, 27157, USA
| | - James B. Daunais
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston Salem, NC, 27157, USA
| | - Richard S. Van
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, 73019, USA
| | - Yihan Shao
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, 73019, USA
| | - Yuqin Wang
- Institute of Life Science, Swansea University Medical School, SA2 8PP Swansea, Wales, United Kingdom
| | - Ming-Rong Zhang
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| | - Catherine Gebhard
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Irina Pikuleva
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Allan I. Levey
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - William J. Griffiths
- Institute of Life Science, Swansea University Medical School, SA2 8PP Swansea, Wales, United Kingdom
| | - Steven H. Liang
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA
- Emory University, Department of Radiology and Imaging Sciences, 1364 Clifton Rd, Atlanta, GA 30322, USA
| |
Collapse
|
12
|
Ma C, Chung DJ, Abramson D, Langley DR, Thayer KM. Mutagenic Activation of Glutathione Peroxidase-4: Approaches toward Rational Design of Allosteric Drugs. ACS OMEGA 2022; 7:29587-29597. [PMID: 36061715 PMCID: PMC9434792 DOI: 10.1021/acsomega.2c01289] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 07/22/2022] [Indexed: 06/15/2023]
Abstract
Glutathione peroxidase 4 (GPX4) reduces lipid hydroperoxides in lipid membranes, effectively inhibiting iron-dependent cell death or ferroptosis. The upregulation of the enzyme by the mutations at residues D21 and D23 has been suggested to be associated with higher protein activity, which confers more protection against neurodegenerative diseases such as Alzheimer's, Parkinson's, and Huntington's diseases. Therefore, it has become an attractive target for treating and preventing neurodegenerative diseases. However, identifying means of mimicking the beneficial effects of these mutations distant from the active site constitutes a formidable challenge in moving toward therapeutics. In this study, we explore using molecular dynamics simulations to computationally map the conformational and energetic landscape of the wild-type GPX4 protein and three mutant variants to identify the allosteric networks of the enzyme. We present the conformational dynamic profile providing the desired signature behavior of the enzyme. We also discuss the implications of these findings for drug design efforts.
Collapse
Affiliation(s)
- Chunyue Ma
- Department
of Mathematics & Computer Science, Wesleyan
University, Middletown, Connecticut 06459, United States
| | - Daniel J. Chung
- Department
of Chemistry, Wesleyan University, Middletown, Connecticut 06459, United States
- Molecular
Biophysics Program, Wesleyan University, Middletown, Connecticut 06459, United States
| | - Dylan Abramson
- Department
of Mathematics & Computer Science, Wesleyan
University, Middletown, Connecticut 06459, United States
| | - David R. Langley
- Department
of Chemistry, Wesleyan University, Middletown, Connecticut 06459, United States
- Molecular
Biophysics Program, Wesleyan University, Middletown, Connecticut 06459, United States
- Arvinas
Inc., New Haven, Connecticut 06511, United States
| | - Kelly M. Thayer
- Department
of Mathematics & Computer Science, Wesleyan
University, Middletown, Connecticut 06459, United States
- Department
of Chemistry, Wesleyan University, Middletown, Connecticut 06459, United States
- Molecular
Biophysics Program, Wesleyan University, Middletown, Connecticut 06459, United States
| |
Collapse
|
13
|
Szlasa W, Janicka N, Sauer N, Michel O, Nowak B, Saczko J, Kulbacka J. Chemotherapy and Physical Therapeutics Modulate Antigens on Cancer Cells. Front Immunol 2022; 13:889950. [PMID: 35874714 PMCID: PMC9299262 DOI: 10.3389/fimmu.2022.889950] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 06/06/2022] [Indexed: 12/29/2022] Open
Abstract
Cancer cells possess specific properties, such as multidrug resistance or unlimited proliferation potential, due to the presence of specific proteins on their cell membranes. The release of proliferation-related proteins from the membrane can evoke a loss of adaptive ability in cancer cells and thus enhance the effects of anticancer therapy. The upregulation of cancer-specific membrane antigens results in a better outcome of immunotherapy. Moreover, cytotoxic T-cells may also become more effective when stimulated ex-vivo toward the anticancer response. Therefore, the modulation of membrane proteins may serve as an interesting attempt in anticancer therapy. The presence of membrane antigens relies on various physical factors such as temperature, exposure to radiation, or drugs. Therefore, changing the tumor microenvironment conditions may lead to cancer cells becoming sensitized to subsequent therapy. This paper focuses on the therapeutic approaches modulating membrane antigens and enzymes in anticancer therapy. It aims to analyze the possible methods for modulating the antigens, such as pharmacological treatment, electric field treatment, photodynamic reaction, treatment with magnetic field or X-ray radiation. Besides, an overview of the effects of chemotherapy and immunotherapy on the immunophenotype of cancer cells is presented. Finally, the authors review the clinical trials that involved the modulation of cell immunophenotype in anticancer therapy.
Collapse
Affiliation(s)
- Wojciech Szlasa
- Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Natalia Janicka
- Faculty of Pharmacy, Wroclaw Medical University, Wroclaw, Poland
| | - Natalia Sauer
- Faculty of Pharmacy, Wroclaw Medical University, Wroclaw, Poland
| | - Olga Michel
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Wroclaw, Poland
| | - Bernadetta Nowak
- Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Jolanta Saczko
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Wroclaw, Poland
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Wroclaw, Poland
| |
Collapse
|
14
|
Cholesterol and its reciprocal association with prion infection. Cell Tissue Res 2022; 392:235-246. [PMID: 35821439 DOI: 10.1007/s00441-022-03669-y] [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: 04/07/2022] [Accepted: 07/04/2022] [Indexed: 11/02/2022]
Abstract
Prion diseases are incurable, infectious and fatal neurodegenerative diseases that affect both humans and animals. The pathogenesis of prion disease involves the misfolding of the cellular prion protein, PrPC, to a disease-causing conformation, PrPSc, in the brain. The exact mechanism of conversion of PrPC to PrPSc is not clear; however, there are numerous studies supporting that this process of misfolding requires the association of PrPC with lipid raft domains of the plasma membrane. An increase in the cellular cholesterol content with prion infection has been observed in both in vivo and in vitro studies. As cholesterol is critical for the formation of lipid rafts, on the one hand, this increase may be related to, or aiding in, the process of prion conversion. On the other hand, increased cholesterol levels may affect neuronal viability. Here, we discuss current literature on the underlying mechanisms and potential consequences of elevated neuronal cholesterol in prion infection and advancements in prion disease therapeutics targeting brain cholesterol homeostasis.
Collapse
|
15
|
Mast N, Li Y, Pikuleva IA. Increased Acetylcholine Levels and Other Brain Effects in 5XFAD Mice after Treatment with 8,14-Dihydroxy Metabolite of Efavirenz. Int J Mol Sci 2022; 23:ijms23147669. [PMID: 35887013 PMCID: PMC9317559 DOI: 10.3390/ijms23147669] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/06/2022] [Accepted: 07/09/2022] [Indexed: 02/08/2023] Open
Abstract
Efavirenz (EFV), an FDA-approved anti-HIV drug, has off-target binding to CYP46A1, the CNS enzyme which converts cholesterol to 24-hydroxycholesterol. At small doses, EFV allosterically activates CYP46A1 in mice and humans and mitigates some of the Alzheimer's disease manifestations in 5XFAD mice, an animal model. Notably, in vitro, all phase 1 EFV hydroxymetabolites activate CYP46A1 as well and bind either to the allosteric site for EFV, neurotransmitters or both. Herein, we treated 5XFAD mice with 8,14-dihydroxyEFV, the binder to the neurotransmitter allosteric site, which elicits the highest CYP46A1 activation in vitro. We found that treated animals of both sexes had activation of CYP46A1 and cholesterol turnover in the brain, decreased content of the amyloid beta 42 peptide, increased levels of acetyl-CoA and acetylcholine, and altered expression of the brain marker proteins. In addition, male mice had improved performance in the Barnes Maze test and increased expression of the acetylcholine-related genes. This work expands our knowledge of the beneficial CYP46A1 activation effects and demonstrates that 8,14-dihydroxyEFV crosses the blood-brain barrier and has therapeutic potential as a CYP46A1 activator.
Collapse
|
16
|
Mast N, Fotinich A, Pikuleva IA. The Hydroxylation Position Rather than Chirality Determines How Efavirenz Metabolites Activate Cytochrome P450 46A1 In Vitro. Drug Metab Dispos 2022; 50:923-930. [PMID: 35489779 PMCID: PMC11022896 DOI: 10.1124/dmd.122.000874] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 04/05/2022] [Indexed: 11/22/2022] Open
Abstract
(S)-Efavirenz (EFV) is a reverse transcriptase inhibitor and an antiviral drug. In addition, (S)-EFV can interact off target with CYP46A1, the major cholesterol hydroxylating enzyme in the mammalian brain, and allosterically activate CYP46A1 at a small dose in mice and humans. Studies with purified CYP46A1 identified two allosteric sites on the enzyme surface, one for (S)-EFV and the second site for L-glutamate (Glu), a neurotransmitter that also activates CYP46A1 either alone or in the presence of (S)-EFV. Previously, we found that racemic (rac)-7-hydroxyefavirenz, (rac)-8-hydroxyefavirenz, (S)-8-hydroxyefavirenz, and (rac)-8,14-dihydroxyefavirenz, compounds with the hydroxylation positions corresponding to the metabolism of (S)-EFV in the liver, activated CYP46A1 in vitro. Yet, these compounds differed from (S)-EFV in how they allosterically interacted with CYP46A1. Herein, we further characterized (rac)-7-hydroxyefavirenz, (rac)-8-hydroxyefavirenz, (S)-8-hydroxyefavirenz, and (rac)-8,14-dihydroxyefavirenz, and, in addition, (R)-EFV, (S)-7-hydroxyefavirenz, (rac)-7,8-dihydroxyefavirenz, (S)-7,8-dihydroxyefavirenz, and (S)-8,14-dihydroxyefavirenz for activation and binding to CYP46A1 in vitro. We found that the spatial configuration of all tested compounds neither affected the CYP46A1 activation nor the sites of binding to CYP46A1. Yet, the hydroxylation position determined whether the hydroxylated metabolite interacted with the allosteric site for (S)-EFV [(R)-EFV, (rac)-7,8-dihydroxyefavirenz, and (S)-7,8-dihydroxyefavirenz], L-Glu [(rac)- and (S)-8,14-dihydroxyefavirenz], or both [(rac)-7-hydroxyefavirenz, (S)-7-hydroxyefavirenz, (rac)-8-hydroxyefavirenz, and (S)-8-hydroxyefavirenz]. This difference in binding to the allosteric sites determined, in turn, how CYP46A1 activity was changed in the coincubations with (S)-EFV and either its metabolite or L-Glu. The results suggest EFV metabolites that could be more potent for CYP46A1 activation in vivo than (S)-EFV. SIGNIFICANCE STATEMENT: This study found that not only efavirenz but also all its hydroxylated metabolites allosterically activate CYP46A1 in vitro. The enzyme activation depended on the hydroxylation position but not the metabolite spatial configuration and involved either one or two allosteric sites-for efavirenz, L-glutamate, or both. The results suggest that the hydroxylated efavirenz metabolites may differ from efavirenz in how they interact with the CYP46A1 allosteric and active sites.
Collapse
Affiliation(s)
- Natalia Mast
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, Ohio
| | - Anna Fotinich
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, Ohio
| | - Irina A Pikuleva
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
17
|
Shi N, Zheng Q, Zhang H. Molecular Basis of the Recognition of Cholesterol by Cytochrome P450 46A1 along the Major Access Tunnel. ACS Chem Neurosci 2022; 13:1526-1533. [PMID: 35438962 DOI: 10.1021/acschemneuro.1c00866] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
CYP46A1 is an important potential target for the treatment of Alzheimer's disease (AD), which is the most common neurodegenerative disease among older individuals. However, the binding mechanism between CYP46A1 and substrate cholesterol (CH) has not been clarified and will not be conducive to the research of relevant drug molecules. In this study, we integrated molecular docking, molecular dynamics (MD) simulations, and adaptive steered MD simulations to explore the recognition and binding mechanism of CYP46A1 with CH. Two key factors affecting the interaction between CH and CYP46A1 are determined: one is a hydrophobic cavity formed by seven hydrophobic residues (F80, Y109, L112, I222, W368, F371, and T475), which provides nonpolar interactions to stabilize CH, and the other is a hydrogen bond formed by H81 and CH, which ensures the binding direction of CH. In addition, the tunnel analysis results show that tunnel 2a is identified as the primary pathway of CH. The entry of CH induces tunnel 2e to close and tunnel w to open. Our results may provide effective clues for the design of drugs based on the substrate for AD and improve our understanding of the structure-function of CYP46A1.
Collapse
Affiliation(s)
- Na Shi
- Institute of Theoretical Chemistry, College of Chemistry, Jilin University, Changchun 130023, China
| | - Qingchuan Zheng
- Institute of Theoretical Chemistry, College of Chemistry, Jilin University, Changchun 130023, China
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, Jilin University, Changchun 130023, China
| | - Hongxing Zhang
- Institute of Theoretical Chemistry, College of Chemistry, Jilin University, Changchun 130023, China
| |
Collapse
|
18
|
Jenkins AJ, Grant MB, Busik JV. Lipids, hyperreflective crystalline deposits and diabetic retinopathy: potential systemic and retinal-specific effect of lipid-lowering therapies. Diabetologia 2022; 65:587-603. [PMID: 35149880 PMCID: PMC9377536 DOI: 10.1007/s00125-022-05655-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 12/14/2021] [Indexed: 12/19/2022]
Abstract
The metabolically active retina obtains essential lipids by endogenous biosynthesis and from the systemic circulation. Clinical studies provide limited and sometimes conflicting evidence as to the relationships between circulating lipid levels and the development and progression of diabetic retinopathy in people with diabetes. Cardiovascular-system-focused clinical trials that also evaluated some retinal outcomes demonstrate the potential protective power of lipid-lowering therapies in diabetic retinopathy and some trials with ocular primary endpoints are in progress. Although triacylglycerol-lowering therapies with fibrates afforded some protection against diabetic retinopathy, the effect was independent of changes in traditional blood lipid classes. While systemic LDL-cholesterol lowering with statins did not afford protection against diabetic retinopathy in most clinical trials, and none of the trials focused on retinopathy as the main outcome, data from very large database studies suggest the possible effectiveness of statins. Potential challenges in these studies are discussed, including lipid-independent effects of fibrates and statins, modified lipoproteins and retinal-specific effects of lipid-lowering drugs. Dysregulation of retinal-specific cholesterol metabolism leading to retinal cholesterol accumulation and potential formation of cholesterol crystals are also addressed.
Collapse
Affiliation(s)
- Alicia J Jenkins
- NHMRC Clinical Trials Centre, The University of Sydney, Sydney, NSW, Australia
| | - Maria B Grant
- Department of Ophthalmology and Vision Science, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Julia V Busik
- Department of Physiology, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
19
|
James EI, Murphree TA, Vorauer C, Engen JR, Guttman M. Advances in Hydrogen/Deuterium Exchange Mass Spectrometry and the Pursuit of Challenging Biological Systems. Chem Rev 2021; 122:7562-7623. [PMID: 34493042 PMCID: PMC9053315 DOI: 10.1021/acs.chemrev.1c00279] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
![]()
Solution-phase hydrogen/deuterium
exchange (HDX) coupled to mass
spectrometry (MS) is a widespread tool for structural analysis across
academia and the biopharmaceutical industry. By monitoring the exchangeability
of backbone amide protons, HDX-MS can reveal information about higher-order
structure and dynamics throughout a protein, can track protein folding
pathways, map interaction sites, and assess conformational states
of protein samples. The combination of the versatility of the hydrogen/deuterium
exchange reaction with the sensitivity of mass spectrometry has enabled
the study of extremely challenging protein systems, some of which
cannot be suitably studied using other techniques. Improvements over
the past three decades have continually increased throughput, robustness,
and expanded the limits of what is feasible for HDX-MS investigations.
To provide an overview for researchers seeking to utilize and derive
the most from HDX-MS for protein structural analysis, we summarize
the fundamental principles, basic methodology, strengths and weaknesses,
and the established applications of HDX-MS while highlighting new
developments and applications.
Collapse
Affiliation(s)
- Ellie I James
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Taylor A Murphree
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Clint Vorauer
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - John R Engen
- Department of Chemistry & Chemical Biology, Northeastern University, Boston, Massachusetts 02115, United States
| | - Miklos Guttman
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington 98195, United States
| |
Collapse
|
20
|
Pikuleva IA, Cartier N. Cholesterol Hydroxylating Cytochrome P450 46A1: From Mechanisms of Action to Clinical Applications. Front Aging Neurosci 2021; 13:696778. [PMID: 34305573 PMCID: PMC8297829 DOI: 10.3389/fnagi.2021.696778] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 06/15/2021] [Indexed: 11/24/2022] Open
Abstract
Cholesterol, an essential component of the brain, and its local metabolism are involved in many neurodegenerative diseases. The blood-brain barrier is impermeable to cholesterol; hence, cholesterol homeostasis in the central nervous system represents a balance between in situ biosynthesis and elimination. Cytochrome P450 46A1 (CYP46A1), a central nervous system-specific enzyme, converts cholesterol to 24-hydroxycholesterol, which can freely cross the blood-brain barrier and be degraded in the liver. By the dual action of initiating cholesterol efflux and activating the cholesterol synthesis pathway, CYP46A1 is the key enzyme that ensures brain cholesterol turnover. In humans and mouse models, CYP46A1 activity is altered in Alzheimer’s and Huntington’s diseases, spinocerebellar ataxias, glioblastoma, and autism spectrum disorders. In mouse models, modulations of CYP46A1 activity mitigate the manifestations of Alzheimer’s, Huntington’s, Nieman-Pick type C, and Machao-Joseph (spinocerebellar ataxia type 3) diseases as well as amyotrophic lateral sclerosis, epilepsy, glioblastoma, and prion infection. Animal studies revealed that the CYP46A1 activity effects are not limited to cholesterol maintenance but also involve critical cellular pathways, like gene transcription, endocytosis, misfolded protein clearance, vesicular transport, and synaptic transmission. How CYP46A1 can exert central control of such essential brain functions is a pressing question under investigation. The potential therapeutic role of CYP46A1, demonstrated in numerous models of brain disorders, is currently being evaluated in early clinical trials. This review summarizes the past 70 years of research that has led to the identification of CYP46A1 and brain cholesterol homeostasis as powerful therapeutic targets for severe pathologies of the CNS.
Collapse
Affiliation(s)
- Irina A Pikuleva
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH, United States
| | - Nathalie Cartier
- NeuroGenCell, Paris Brain Institute, ICM, Inserm U 1127, CNRS UMR 7225, Sorbonne Université, Paris, France
| |
Collapse
|
21
|
Souza SA, Held A, Lu WJ, Drouhard B, Avila B, Leyva-Montes R, Hu M, Miller BR, Ng HL. Mechanisms of allosteric and mixed mode aromatase inhibitors. RSC Chem Biol 2021; 2:892-905. [PMID: 34458816 PMCID: PMC8341375 DOI: 10.1039/d1cb00046b] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 03/17/2021] [Indexed: 11/21/2022] Open
Abstract
Aromatase (CYP19) catalyzes the last biosynthetic step of estrogens in mammals and is a primary drug target for hormone-related breast cancer. However, treatment with aromatase inhibitors is often associated with adverse effects and drug resistance. In this study, we used virtual screening targeting a predicted cytochrome P450 reductase binding site on aromatase to discover four novel non-steroidal aromatase inhibitors. The inhibitors have potencies comparable to the noncompetitive tamoxifen metabolite, endoxifen. Our two most potent inhibitors, AR11 and AR13, exhibit both mixed-type and competitive-type inhibition. The cytochrome P450 reductase-CYP19 coupling interface likely acts as a transient binding site. Our modeling shows that our inhibitors bind better at different sites near the catalytic site. Our results predict the location of multiple ligand binding sites on aromatase. The combination of modeling and experimental results supports the important role of the reductase binding interface as a low affinity, promiscuous ligand binding site. Our new inhibitors may be useful as alternative chemical scaffolds that may show different adverse effects profiles than current clinically used aromatase inhibitors.
Collapse
Affiliation(s)
- Samson A Souza
- Department of Biochemistry and Molecular Biophysics, Kansas State University Manhattan KS USA
| | - Abby Held
- Department of Chemistry, Truman State University Kirksville MO USA
| | - Wenjie J Lu
- Department of Chemistry, University of Hawai'i at Mānoa Honolulu HI USA
| | - Brendan Drouhard
- Department of Biochemistry and Molecular Biophysics, Kansas State University Manhattan KS USA
| | - Bryant Avila
- Department of Biochemistry and Molecular Biophysics, Kansas State University Manhattan KS USA
| | - Raul Leyva-Montes
- Department of Biochemistry and Molecular Biophysics, Kansas State University Manhattan KS USA
| | - Michelle Hu
- Department of Chemistry, University of Hawai'i at Mānoa Honolulu HI USA
| | - Bill R Miller
- Department of Chemistry, Truman State University Kirksville MO USA
| | - Ho Leung Ng
- Department of Biochemistry and Molecular Biophysics, Kansas State University Manhattan KS USA
| |
Collapse
|
22
|
Oral administration of repurposed drug targeting Cyp46A1 increases survival times of prion infected mice. Acta Neuropathol Commun 2021; 9:58. [PMID: 33795005 PMCID: PMC8017635 DOI: 10.1186/s40478-021-01162-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 03/17/2021] [Indexed: 01/12/2023] Open
Abstract
Prion diseases are fatal, infectious, and incurable neurodegenerative disorders caused by misfolding of the cellular prion protein (PrPC) into the infectious isoform (PrPSc). In humans, there are sporadic, genetic and infectious etiologies, with sporadic Creutzfeldt-Jakob disease (sCJD) being the most common form. Currently, no treatment is available for prion diseases. Cellular cholesterol is known to impact prion conversion, which in turn results in an accumulation of cholesterol in prion-infected neurons. The major elimination of brain cholesterol is achieved by the brain specific enzyme, cholesterol 24-hydroxylase (CYP46A1). Cyp46A1 converts cholesterol into 24(S)-hydroxycholesterol, a membrane-permeable molecule that exits the brain. We have demonstrated for the first time that Cyp46A1 levels are reduced in the brains of prion-infected mice at advanced disease stage, in prion-infected neuronal cells and in post-mortem brains of sCJD patients. We have employed the Cyp46A1 activator efavirenz (EFV) for treatment of prion-infected neuronal cells and mice. EFV is an FDA approved anti-HIV medication effectively crossing the blood brain barrier and has been used for decades to chronically treat HIV patients. EFV significantly mitigated PrPSc propagation in prion-infected cells while preserving physiological PrPC and lipid raft integrity. Notably, oral administration of EFV treatment chronically at very low dosage starting weeks to months after intracerebral prion inoculation of mice significantly prolonged the lifespan of animals. In summary, our results suggest that Cyp46A1 as a novel therapeutic target and that its activation through repurposing the anti-retroviral medication EFV might be valuable treatment approach for prion diseases.
Collapse
|
23
|
Sodero AO. 24S-hydroxycholesterol: Cellular effects and variations in brain diseases. J Neurochem 2020; 157:899-918. [PMID: 33118626 DOI: 10.1111/jnc.15228] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 10/13/2020] [Accepted: 10/17/2020] [Indexed: 12/12/2022]
Abstract
The adult brain exhibits a characteristic cholesterol homeostasis, with low synthesis rate and active catabolism. Brain cholesterol turnover is possible thanks to the action of the enzyme cytochrome P450 46A1 (CYP46A1) or 24-cholesterol hydroxylase, that transforms cholesterol into 24S-hydroxycholesterol (24S-HC). But before crossing the blood-brain barrier (BBB), this oxysterol, that is the most abundant in the brain, can act locally, affecting the functioning of neurons, astrocytes, oligodendrocytes, and vascular cells. The first part of this review addresses different aspects of 24S-HC production and elimination from the brain. The second part concentrates in the effects of 24S-HC at the cellular level, describing how this oxysterol affects cell viability, amyloid β production, neurotransmission, and transcriptional activity. Finally, the role of 24S-HC in Alzheimer, Huntington and Parkinson diseases, multiple sclerosis and amyotrophic lateral sclerosis, as well as the possibility of using this oxysterol as predictive and/or evolution biomarker in different brain disorders is discussed.
Collapse
Affiliation(s)
- Alejandro O Sodero
- Institute of Biomedical Research (BIOMED), Pontifical Catholic University of Argentina (UCA) and National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| |
Collapse
|
24
|
Paço L, Zarate-Perez F, Clouser AF, Atkins WM, Hackett JC. Dynamics and Mechanism of Binding of Androstenedione to Membrane-Associated Aromatase. Biochemistry 2020; 59:2999-3009. [DOI: 10.1021/acs.biochem.0c00460] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Lorela Paço
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington 98195-7610, United States
| | - Francisco Zarate-Perez
- Department of Physiology and Biophysics and Massey Cancer Center, School of Medicine, Virginia Commonwealth University, Richmond, Virginia 23298-0035, United States
| | - Amanda F. Clouser
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington 98195-7610, United States
| | - William M. Atkins
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington 98195-7610, United States
| | - John C. Hackett
- Department of Physiology and Biophysics and Massey Cancer Center, School of Medicine, Virginia Commonwealth University, Richmond, Virginia 23298-0035, United States
| |
Collapse
|
25
|
Perinatal exposure of rats to the HIV drug efavirenz affects medial prefrontal cortex cytoarchitecture. Biochem Pharmacol 2020; 178:114050. [PMID: 32446887 DOI: 10.1016/j.bcp.2020.114050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 05/19/2020] [Indexed: 12/29/2022]
Abstract
Efavirenz (EFV) is used for antiretroviral treatment of HIV infection, and successfully inhibits viral replication and mother-to-child transmission of HIV during pregnancy and childbirth. Unfortunately, the drug induces neuropsychiatric symptoms such as anxiety and depressed mood and potentially affects cognitive performance. EFV acts on, among others, the serotonin transporter and serotonin receptors that are expressed in the developing brain. Yet, how perinatal EFV exposure affects brain cytoarchitecture remains unclear. Here, we exposed pregnant and lactating rats to EFV, and examined in the medial prefrontal cortex (mPFC) of their adult offspring the effects of the maternal EFV exposure on cortical architecture. We observed a significant decrease in the number of cells, mainly mature neurons, in the infra/prelimbic and cingulate cortices of adult offspring. Next, we found an altered cortical cytoarchitecture characterized by a significant reduction in deep- and superficial-layer cells. This was accompanied by a sharp increase in programmed cell death, as we identified a significantly higher number of cleaved Caspase-3-positive cells. Finally, the serotonergic and dopaminergic innervation of the mPFC subdomains was increased. Thus, the perinatal exposure to EFV provoked in the mPFC of adult offspring cell death, significant changes in cytoarchitecture, and disturbances in serotonergic and dopaminergic innervation. Our results are important in the light of EFV treatment of HIV-positive pregnant women, and its effect on brain development and cognitive behavior.
Collapse
|
26
|
Jeffreys LN, Pacholarz KJ, Johannissen LO, Girvan HM, Barran PE, Voice MW, Munro AW. Characterization of the structure and interactions of P450 BM3 using hybrid mass spectrometry approaches. J Biol Chem 2020; 295:7595-7607. [PMID: 32303637 PMCID: PMC7261786 DOI: 10.1074/jbc.ra119.011630] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 04/02/2020] [Indexed: 01/08/2023] Open
Abstract
The cytochrome P450 monooxygenase P450 BM3 (BM3) is a biotechnologically important and versatile enzyme capable of producing important compounds such as the medical drugs pravastatin and artemether, and the steroid hormone testosterone. BM3 is a natural fusion enzyme comprising two major domains: a cytochrome P450 (heme-binding) catalytic domain and a NADPH-cytochrome P450 reductase (CPR) domain containing FAD and FMN cofactors in distinct domains of the CPR. A crystal structure of full-length BM3 enzyme is not available in its monomeric or catalytically active dimeric state. In this study, we provide detailed insights into the protein-protein interactions that occur between domains in the BM3 enzyme and characterize molecular interactions within the BM3 dimer by using several hybrid mass spectrometry (MS) techniques, namely native ion mobility MS (IM-MS), collision-induced unfolding (CIU), and hydrogen-deuterium exchange MS (HDX-MS). These methods enable us to probe the structure, stoichiometry, and domain interactions in the ∼240 kDa BM3 dimeric complex. We obtained high-sequence coverage (88–99%) in the HDX-MS experiments for full-length BM3 and its component domains in both the ligand-free and ligand-bound states. We identified important protein interaction sites, in addition to sites corresponding to heme-CPR domain interactions at the dimeric interface. These findings bring us closer to understanding the structure and catalytic mechanism of P450 BM3.
Collapse
Affiliation(s)
- Laura N Jeffreys
- The Manchester Institute of Biotechnology, School of Natural Sciences, Department of Chemistry, The University of Manchester, 131 Princess Street, Manchester M1 7DN, United Kingdom.,Manchester Synthetic Biology Research Centre for Fine and Speciality Chemicals (SYNBIOCHEM), The University of Manchester, 131 Princess Street, Manchester M1 7DN, United Kingdom
| | - Kamila J Pacholarz
- Michael Barber Centre for Collaborative Mass Spectrometry, Manchester Institute of Biotechnology, The University of Manchester, 131 Princess Street, Manchester M1 7DN, United Kingdom
| | - Linus O Johannissen
- The Manchester Institute of Biotechnology, School of Natural Sciences, Department of Chemistry, The University of Manchester, 131 Princess Street, Manchester M1 7DN, United Kingdom.,Manchester Synthetic Biology Research Centre for Fine and Speciality Chemicals (SYNBIOCHEM), The University of Manchester, 131 Princess Street, Manchester M1 7DN, United Kingdom
| | - Hazel M Girvan
- The Manchester Institute of Biotechnology, School of Natural Sciences, Department of Chemistry, The University of Manchester, 131 Princess Street, Manchester M1 7DN, United Kingdom.,Manchester Synthetic Biology Research Centre for Fine and Speciality Chemicals (SYNBIOCHEM), The University of Manchester, 131 Princess Street, Manchester M1 7DN, United Kingdom
| | - Perdita E Barran
- The Manchester Institute of Biotechnology, School of Natural Sciences, Department of Chemistry, The University of Manchester, 131 Princess Street, Manchester M1 7DN, United Kingdom.,Manchester Synthetic Biology Research Centre for Fine and Speciality Chemicals (SYNBIOCHEM), The University of Manchester, 131 Princess Street, Manchester M1 7DN, United Kingdom.,Michael Barber Centre for Collaborative Mass Spectrometry, Manchester Institute of Biotechnology, The University of Manchester, 131 Princess Street, Manchester M1 7DN, United Kingdom
| | - Michael W Voice
- Cypex Ltd., 6 Tom McDonald Avenue, Dundee, DD2 1NH, United Kingdom
| | - Andrew W Munro
- The Manchester Institute of Biotechnology, School of Natural Sciences, Department of Chemistry, The University of Manchester, 131 Princess Street, Manchester M1 7DN, United Kingdom .,Manchester Synthetic Biology Research Centre for Fine and Speciality Chemicals (SYNBIOCHEM), The University of Manchester, 131 Princess Street, Manchester M1 7DN, United Kingdom
| |
Collapse
|
27
|
Han M, Wang S, Yang N, Wang X, Zhao W, Saed HS, Daubon T, Huang B, Chen A, Li G, Miletic H, Thorsen F, Bjerkvig R, Li X, Wang J. Therapeutic implications of altered cholesterol homeostasis mediated by loss of CYP46A1 in human glioblastoma. EMBO Mol Med 2020; 12:e10924. [PMID: 31777202 PMCID: PMC6949512 DOI: 10.15252/emmm.201910924] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 10/19/2019] [Accepted: 10/23/2019] [Indexed: 01/04/2023] Open
Abstract
Dysregulated cholesterol metabolism is a hallmark of many cancers, including glioblastoma (GBM), but its role in disease progression is not well understood. Here, we identified cholesterol 24-hydroxylase (CYP46A1), a brain-specific enzyme responsible for the elimination of cholesterol through the conversion of cholesterol into 24(S)-hydroxycholesterol (24OHC), as one of the most dramatically dysregulated cholesterol metabolism genes in GBM. CYP46A1 was significantly decreased in GBM samples compared with normal brain tissue. A reduction in CYP46A1 expression was associated with increasing tumour grade and poor prognosis in human gliomas. Ectopic expression of CYP46A1 suppressed cell proliferation and in vivo tumour growth by increasing 24OHC levels. RNA-seq revealed that treatment of GBM cells with 24OHC suppressed tumour growth through regulation of LXR and SREBP signalling. Efavirenz, an activator of CYP46A1 that is known to penetrate the blood-brain barrier, inhibited GBM growth in vivo. Our findings demonstrate that CYP46A1 is a critical regulator of cellular cholesterol in GBM and that the CYP46A1/24OHC axis is a potential therapeutic target.
Collapse
Affiliation(s)
- Mingzhi Han
- Shandong Key Laboratory of Brain Function RemodelingDepartment of NeurosurgeryQilu Hospital of Shandong University and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinanChina
- Department of BiomedicineUniversity of BergenBergenNorway
| | - Shuai Wang
- Shandong Key Laboratory of Brain Function RemodelingDepartment of NeurosurgeryQilu Hospital of Shandong University and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinanChina
| | - Ning Yang
- Shandong Key Laboratory of Brain Function RemodelingDepartment of NeurosurgeryQilu Hospital of Shandong University and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinanChina
| | - Xu Wang
- Shandong Key Laboratory of Brain Function RemodelingDepartment of NeurosurgeryQilu Hospital of Shandong University and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinanChina
| | - Wenbo Zhao
- Shandong Key Laboratory of Brain Function RemodelingDepartment of NeurosurgeryQilu Hospital of Shandong University and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinanChina
| | | | - Thomas Daubon
- INSERM U1029Institut Nationale de la Santé et de la Recherche MédicalePessacFrance
- University BordeauxPessacFrance
| | - Bin Huang
- Shandong Key Laboratory of Brain Function RemodelingDepartment of NeurosurgeryQilu Hospital of Shandong University and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinanChina
| | - Anjing Chen
- Shandong Key Laboratory of Brain Function RemodelingDepartment of NeurosurgeryQilu Hospital of Shandong University and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinanChina
- School of MedicineShandong UniversityJinanChina
| | - Gang Li
- Shandong Key Laboratory of Brain Function RemodelingDepartment of NeurosurgeryQilu Hospital of Shandong University and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinanChina
| | - Hrvoje Miletic
- Department of BiomedicineUniversity of BergenBergenNorway
- Department of PathologyHaukeland University HospitalBergenNorway
| | - Frits Thorsen
- Department of BiomedicineUniversity of BergenBergenNorway
- Department of BiomedicineThe Molecular Imaging CenterUniversity of BergenBergenNorway
| | - Rolf Bjerkvig
- Department of BiomedicineUniversity of BergenBergenNorway
- NorLux Neuro‐Oncology LaboratoryDepartment of OncologyLuxembourg Institute of HealthLuxembourg CityLuxembourg
| | - Xingang Li
- Shandong Key Laboratory of Brain Function RemodelingDepartment of NeurosurgeryQilu Hospital of Shandong University and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinanChina
| | - Jian Wang
- Shandong Key Laboratory of Brain Function RemodelingDepartment of NeurosurgeryQilu Hospital of Shandong University and Institute of Brain and Brain‐Inspired ScienceShandong UniversityJinanChina
- Department of BiomedicineUniversity of BergenBergenNorway
| |
Collapse
|
28
|
Amyloid-β-independent regulators of tau pathology in Alzheimer disease. Nat Rev Neurosci 2019; 21:21-35. [PMID: 31780819 DOI: 10.1038/s41583-019-0240-3] [Citation(s) in RCA: 316] [Impact Index Per Article: 63.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2019] [Indexed: 12/12/2022]
Abstract
The global epidemic of Alzheimer disease (AD) is worsening, and no approved treatment can revert or arrest progression of this disease. AD pathology is characterized by the accumulation of amyloid-β (Aβ) plaques and tau neurofibrillary tangles in the brain. Genetic data, as well as autopsy and neuroimaging studies in patients with AD, indicate that Aβ plaque deposition precedes cortical tau pathology. Because Aβ accumulation has been considered the initial insult that drives both the accumulation of tau pathology and tau-mediated neurodegeneration in AD, the development of AD therapeutics has focused mostly on removing Aβ from the brain. However, striking preclinical evidence from AD mouse models and patient-derived human induced pluripotent stem cell models indicates that tau pathology can progress independently of Aβ accumulation and arises downstream of genetic risk factors for AD and aberrant metabolic pathways. This Review outlines novel insights from preclinical research that implicate apolipoprotein E, the endocytic system, cholesterol metabolism and microglial activation as Aβ-independent regulators of tau pathology. These factors are discussed in the context of emerging findings from clinical pathology, functional neuroimaging and other approaches in humans. Finally, we discuss the implications of these new insights for current Aβ-targeted strategies and highlight the emergence of novel therapeutic strategies that target processes upstream of both Aβ and tau.
Collapse
|
29
|
Mast N, Verwilst P, Wilkey CJ, Guengerich FP, Pikuleva IA. In Vitro Activation of Cytochrome P450 46A1 (CYP46A1) by Efavirenz-Related Compounds. J Med Chem 2019; 63:6477-6488. [PMID: 31617715 PMCID: PMC7226586 DOI: 10.1021/acs.jmedchem.9b01383] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
![]()
Cytochrome P450 46A1 (CYP46A1) is a central nervous system-specific
enzyme, which catalyzes cholesterol 24-hydroxylation. Currently CYP46A1
is being evaluated in a clinical trial for activation by small doses
of the anti-HIV drug efavirenz. Eight efavirenz-related compounds
were investigated for CYP46A1 activation in vitro, induction of a
CYP46A1 spectral response, spectral Kd values, interaction with the P450 allosteric sites, and a model
of binding to the enzyme active site. We gained insight into structure–activity
relationships of efavirenz for CYP46A1 activation and found that the
investigated efavirenz primary metabolites are stronger and better
activators of CYP46A1 than efavirenz. We also established that CYP46A1
is activated by racemates and that a conformational-selection mechanism
is operative in CYP46A1. The results suggest structural modifications
of efavirenz to further increase CYP46A1 activation without inhibition
at high compound concentrations. It is possible that not only efavirenz
but its metabolites activate CYP46A1 in vivo.
Collapse
Affiliation(s)
- Natalia Mast
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Peter Verwilst
- Department of Chemistry, Korea University, Seoul 02841, Korea
| | - Clayton J Wilkey
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - F Peter Guengerich
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Irina A Pikuleva
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, Ohio 44106, United States
| |
Collapse
|
30
|
Petrov AM, Pikuleva IA. Cholesterol 24-Hydroxylation by CYP46A1: Benefits of Modulation for Brain Diseases. Neurotherapeutics 2019; 16:635-648. [PMID: 31001737 PMCID: PMC6694357 DOI: 10.1007/s13311-019-00731-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Cholesterol 24-hydroxylation is the major mechanism for cholesterol removal from the brain and the reaction catalyzed by cytochrome P450 46A1 (CYP46A1), a CNS-specific enzyme. This review describes CYP46A1 in the context of cholesterol homeostasis in the brain and summarizes available experimental data on CYP46A1 association with different neurologic diseases, including the mechanisms by which changes in the CYP46A1 activity in the brain could be beneficial for these diseases. The modulation of CYP46A1 activity by genetic and pharmacologic means is also presented along with a brief synopsis of the two clinical trials that evaluate CYP46A1 as a therapeutic target for Alzheimer's disease as well as Dravet and Lennox-Gastaut syndromes.
Collapse
Affiliation(s)
- Alexey M Petrov
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, 2085 Adelbert Rd., Room 303, Cleveland, OH, 44106, USA
| | - Irina A Pikuleva
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, 2085 Adelbert Rd., Room 303, Cleveland, OH, 44106, USA.
| |
Collapse
|
31
|
Petrov AM, Lam M, Mast N, Moon J, Li Y, Maxfield E, Pikuleva IA. CYP46A1 Activation by Efavirenz Leads to Behavioral Improvement without Significant Changes in Amyloid Plaque Load in the Brain of 5XFAD Mice. Neurotherapeutics 2019; 16:710-724. [PMID: 31062296 PMCID: PMC6694340 DOI: 10.1007/s13311-019-00737-0] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Efavirenz, the FDA-approved anti-retroviral medication, is evaluated in the clinical trial in patients with mild cognitive impairment or early dementia due to Alzheimer's disease. Efavirenz is assessed for activation of cytochrome P450 46A1 (CYP46A1), a CNS-specific enzyme that converts cholesterol to 24-hydroxycholesterol. Cholesterol 24-hydroxylation is the major pathway for brain cholesterol removal, and a mechanism that controls brain cholesterol turnover. The present study tested efavirenz on 5XFAD mice (an Alzheimer's model) at a very low daily dose of 0.1 mg/kg body weight. Efavirenz treatment started from three months of age, after amyloid plague appearance, and continued for 6 months. This treatment led to CYP46A1 activation in the brain, enhancement of brain cholesterol turnover, behavioral improvements, reduction in microglia activation but increased astrocyte reactivity. The levels of the soluble and insoluble amyloid 40 and 42 peptides were unchanged while the number and area of the dense core amyloid plaques were slightly decreased. The measurements of the brain levels of several pre- and post-synaptic proteins (Munc13-1, PSD-95, gephyrin, synaptophysin, synapsin-1, and calbindin-D28k) suggested efavirenz effect at the synaptic level. Efavirenz treatment in the present work seems to represent a model of behavioral and other improvements independent of the levels of the amyloid peptides and provides insight into potential outcomes of the future clinical trial.
Collapse
Affiliation(s)
- Alexey M Petrov
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, 2085 Adelbert Rd., Room 303, Cleveland, OH, 44106, USA
| | - Morrie Lam
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, 2085 Adelbert Rd., Room 303, Cleveland, OH, 44106, USA
| | - Natalia Mast
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, 2085 Adelbert Rd., Room 303, Cleveland, OH, 44106, USA
| | - Jean Moon
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, 2085 Adelbert Rd., Room 303, Cleveland, OH, 44106, USA
| | - Yong Li
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, 2085 Adelbert Rd., Room 303, Cleveland, OH, 44106, USA
| | - Erin Maxfield
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, 2085 Adelbert Rd., Room 303, Cleveland, OH, 44106, USA
| | - Irina A Pikuleva
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, 2085 Adelbert Rd., Room 303, Cleveland, OH, 44106, USA.
| |
Collapse
|
32
|
Rendic SP, Peter Guengerich F. Human cytochrome P450 enzymes 5-51 as targets of drugs and natural and environmental compounds: mechanisms, induction, and inhibition - toxic effects and benefits. Drug Metab Rev 2019; 50:256-342. [PMID: 30717606 DOI: 10.1080/03602532.2018.1483401] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cytochrome P450 (P450, CYP) enzymes have long been of interest due to their roles in the metabolism of drugs, pesticides, pro-carcinogens, and other xenobiotic chemicals. They have also been of interest due to their very critical roles in the biosynthesis and metabolism of steroids, vitamins, and certain eicosanoids. This review covers the 22 (of the total of 57) human P450s in Families 5-51 and their substrate selectivity. Furthermore, included is information and references regarding inducibility, inhibition, and (in some cases) stimulation by chemicals. We update and discuss important aspects of each of these 22 P450s and questions that remain open.
Collapse
Affiliation(s)
| | - F Peter Guengerich
- b Department of Biochemistry , Vanderbilt University School of Medicine , Nashville , TN , USA
| |
Collapse
|
33
|
Huff HC, Maroutsos D, Das A. Lipid composition and macromolecular crowding effects on CYP2J2-mediated drug metabolism in nanodiscs. Protein Sci 2019; 28:928-940. [PMID: 30861250 DOI: 10.1002/pro.3603] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 03/11/2019] [Indexed: 01/13/2023]
Abstract
Lipid composition and macromolecular crowding are key external effectors of protein activity and stability whose role varies between different proteins. Therefore, it is imperative to study their effects on individual protein function. CYP2J2 is a membrane-bound cytochrome P450 in the heart involved in the metabolism of fatty acids and xenobiotics. In order to facilitate this metabolism, cytochrome P450 reductase (CPR), transfers electrons to CYP2J2 from NADPH. Herein, we use nanodiscs to show that lipid composition of the membrane bilayer affects substrate metabolism of the CYP2J2-CPR nanodisc (ND) system. Differential effects on both NADPH oxidation and substrate metabolism by CYP2J2-CPR are dependent on the lipid composition. For instance, sphingomyelin containing nanodiscs produced more secondary substrate metabolites than discs of other lipid compositions, implying a possible conformational change leading to processive metabolism. Furthermore, we demonstrate that macromolecular crowding plays a role in the lipid-solubilized CYP2J2-CPR system by increasing the Km and decreasing the Vmax , and effect that is size-dependent. Crowding also affects the CYP2J2-CPR-ND system by decreasing both the Km and Vmax for Dextran-based macromolecular crowding agents, implying an increase in substrate affinity but a lack of metabolism. Finally, protein denaturation studies show that crowding agents destabilize CYP2J2, while the multidomain protein CPR is stabilized. Overall, these studies are the first report on the role of the surrounding lipid environment and macromolecular crowding in modulating enzymatic function of CYP2J2-CPR membrane protein system.
Collapse
Affiliation(s)
- Hannah C Huff
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801
| | - Demetri Maroutsos
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801
| | - Aditi Das
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801.,Beckman Institute for Advanced Science and Technology, Division of Nutritional Science, Neuroscience Program, and Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801.,Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801
| |
Collapse
|
34
|
van der Kant R, Langness VF, Herrera CM, Williams DA, Fong LK, Leestemaker Y, Steenvoorden E, Rynearson KD, Brouwers JF, Helms JB, Ovaa H, Giera M, Wagner SL, Bang AG, Goldstein LSB. Cholesterol Metabolism Is a Druggable Axis that Independently Regulates Tau and Amyloid-β in iPSC-Derived Alzheimer's Disease Neurons. Cell Stem Cell 2019; 24:363-375.e9. [PMID: 30686764 PMCID: PMC6414424 DOI: 10.1016/j.stem.2018.12.013] [Citation(s) in RCA: 203] [Impact Index Per Article: 40.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 09/26/2018] [Accepted: 12/17/2018] [Indexed: 02/07/2023]
Abstract
Genetic, epidemiologic, and biochemical evidence suggests that predisposition to Alzheimer's disease (AD) may arise from altered cholesterol metabolism, although the molecular pathways that may link cholesterol to AD phenotypes are only partially understood. Here, we perform a phenotypic screen for pTau accumulation in AD-patient iPSC-derived neurons and identify cholesteryl esters (CE), the storage product of excess cholesterol, as upstream regulators of Tau early during AD development. Using isogenic induced pluripotent stem cell (iPSC) lines carrying mutations in the cholesterol-binding domain of APP or APP null alleles, we found that while CE also regulate Aβ secretion, the effects of CE on Tau and Aβ are mediated by independent pathways. Efficacy and toxicity screening in iPSC-derived astrocytes and neurons showed that allosteric activation of CYP46A1 lowers CE specifically in neurons and is well tolerated by astrocytes. These data reveal that CE independently regulate Tau and Aβ and identify a druggable CYP46A1-CE-Tau axis in AD.
Collapse
Affiliation(s)
- Rik van der Kant
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University Amsterdam de Boelelaan 1087, 1081 HV Amsterdam, the Netherlands
| | - Vanessa F Langness
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Cheryl M Herrera
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Daniel A Williams
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Lauren K Fong
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Yves Leestemaker
- Oncode Institute and Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, the Netherlands
| | - Evelyne Steenvoorden
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Kevin D Rynearson
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jos F Brouwers
- Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine, Utrecht University Yalelaan 2, 3584 CM Utrecht, the Netherlands
| | - J Bernd Helms
- Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine, Utrecht University Yalelaan 2, 3584 CM Utrecht, the Netherlands
| | - Huib Ovaa
- Oncode Institute and Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, the Netherlands
| | - Martin Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Steven L Wagner
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, USA; Research Biologist, VA San Diego Healthcare System, La Jolla, CA 92161, USA
| | - Anne G Bang
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Lawrence S B Goldstein
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA.
| |
Collapse
|
35
|
Srinivas N, Rosen EP, Gilliland WM, Kovarova M, Remling-Mulder L, De La Cruz G, White N, Adamson L, Schauer AP, Sykes C, Luciw P, Garcia JV, Akkina R, Kashuba ADM. Antiretroviral concentrations and surrogate measures of efficacy in the brain tissue and CSF of preclinical species. Xenobiotica 2018; 49:1192-1201. [PMID: 30346892 DOI: 10.1080/00498254.2018.1539278] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
1. Antiretroviral concentrations in cerebrospinal fluid (CSF) are used as surrogate for brain tissue, although sparse data support this. We quantified antiretrovirals in brain tissue across preclinical models, compared them to CSF, and calculated 90% inhibitory quotients (IQ90) for nonhuman primate (NHP) brain tissue. Spatial distribution of efavirenz was performed by mass-spectrometry imaging (MSI). 2. HIV or RT-SHIV-infected and uninfected animals from two humanized mouse models (hemopoietic-stem cell/RAG2-, n = 36; bone marrow-liver-thymus/BLT, n =13) and an NHP model (rhesus macaque, n =18) were dosed with six antiretrovirals. Brain tissue, CSF (NHPs), and plasma were collected at necropsy. Drug concentrations were measured by LC-MS/MS. Rapid equilibrium dialysis determined protein binding in NHP brain. 3. Brain tissue penetration of most antiretrovirals were >10-fold lower (p < 0.02) in humanized mice than NHPs. NHP CSF concentrations were >13-fold lower (p <0.02) than brain tissue with poor agreement except for efavirenz (r = 0.91, p = 0.001). Despite 97% brain tissue protein binding, efavirenz achieved IQ90>1 in all animals and 2-fold greater white versus gray matter concentration. 4. Brain tissue penetration varied across animal models for all antiretrovirals except raltegravir, and extrapolating brain tissue concentrations between models should be avoided. With the exception of efavirenz, CSF is not a surrogate for brain tissue concentrations.
Collapse
Affiliation(s)
- Nithya Srinivas
- a Eshelman School of Pharmacy , University of North Carolina at Chapel Hill , Chapel Hill , NC , USA
| | - Elias P Rosen
- a Eshelman School of Pharmacy , University of North Carolina at Chapel Hill , Chapel Hill , NC , USA
| | - William M Gilliland
- a Eshelman School of Pharmacy , University of North Carolina at Chapel Hill , Chapel Hill , NC , USA
| | - Martina Kovarova
- b School of Medicine , University of North Carolina at Chapel Hill , Chapel Hill , NC , USA
| | | | - Gabriela De La Cruz
- b School of Medicine , University of North Carolina at Chapel Hill , Chapel Hill , NC , USA
| | - Nicole White
- a Eshelman School of Pharmacy , University of North Carolina at Chapel Hill , Chapel Hill , NC , USA
| | - Lourdes Adamson
- d School of Medicine , University of California at Davis , Davis , CA , USA
| | - Amanda P Schauer
- a Eshelman School of Pharmacy , University of North Carolina at Chapel Hill , Chapel Hill , NC , USA
| | - Craig Sykes
- a Eshelman School of Pharmacy , University of North Carolina at Chapel Hill , Chapel Hill , NC , USA
| | - Paul Luciw
- d School of Medicine , University of California at Davis , Davis , CA , USA
| | - J Victor Garcia
- b School of Medicine , University of North Carolina at Chapel Hill , Chapel Hill , NC , USA
| | - Ramesh Akkina
- c School of Medicine , Colorado State University , Fort Collins , CO , USA
| | - Angela D M Kashuba
- a Eshelman School of Pharmacy , University of North Carolina at Chapel Hill , Chapel Hill , NC , USA
| |
Collapse
|
36
|
Polic V, Sevrioukova IF, Auclair K. Steroid bioconjugation to a CYP3A4 allosteric site and its effect on substrate binding and coupling efficiency. Arch Biochem Biophys 2018; 653:90-96. [PMID: 29958895 PMCID: PMC6450699 DOI: 10.1016/j.abb.2018.06.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 06/11/2018] [Accepted: 06/25/2018] [Indexed: 12/17/2022]
Abstract
Human cytochrome P450 3A4 (CYP3A4) is an important drug metabolizing enzyme involved in a number of drug-drug and food-drug interactions. As such, much effort has been devoted into investigating its mechanism of interaction with ligands. CYP3A4 has one of the highest levels of substrate promiscuity for an enzyme, and can even bind multiple ligands simultaneously. The location and orientation of these ligands depend on the chemical structure and stoichiometry, and are generally poorly understood. In the case of the steroid testosterone, up to three copies of the molecule can associate with the enzyme at once, likely two in the active site and one at a postulated allosteric site. Recently, we demonstrated that steroid bioconjugation at the allosteric site results in an increase in activity of CYP3A4 toward testosterone and 7-benzyloxy-4-trifluoromethylcoumarin oxidation. Here, using the established bioconjugation methodology, we show how steroid bioconjugation at the allosteric site affects the heme spin state, the binding affinity (KS) of CYP3A4 for testosterone, as well as the enzyme coupling efficiency.
Collapse
Affiliation(s)
- Vanja Polic
- Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montreal, Quebec, H3A 0B8, Canada
| | - Irina F Sevrioukova
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, 92697, United States
| | - Karine Auclair
- Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montreal, Quebec, H3A 0B8, Canada.
| |
Collapse
|
37
|
Thayer KM, Galganov JC, Stein AJ. Dependence of prevalence of contiguous pathways in proteins on structural complexity. PLoS One 2017; 12:e0188616. [PMID: 29232711 PMCID: PMC5726733 DOI: 10.1371/journal.pone.0188616] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 11/10/2017] [Indexed: 12/15/2022] Open
Abstract
Allostery is a regulatory mechanism in proteins where an effector molecule binds distal from an active site to modulate its activity. Allosteric signaling may occur via a continuous path of residues linking the active and allosteric sites, which has been suggested by large conformational changes evident in crystal structures. An alternate possibility is that the signal occurs in the realm of ensemble dynamics via an energy landscape change. While the latter was first proposed on theoretical grounds, increasing evidence suggests that such a control mechanism is plausible. A major difficulty for testing the two methods is the ability to definitively determine that a residue is directly involved in allosteric signal transduction. Statistical Coupling Analysis (SCA) is a method that has been successful at predicting pathways, and experimental tests involving mutagenesis or domain substitution provide the best available evidence of signaling pathways. However, ascertaining energetic pathways which need not be contiguous is far more difficult. To date, simple estimates of the statistical significance of a pathway in a protein remain to be established. The focus of this work is to estimate such benchmarks for the statistical significance of contiguous pathways for the null model of selecting residues at random. We found that when 20% of residues in proteins are randomly selected, contiguous pathways at the 6 Å cutoff level were found with success rates of 51% in PDZ, 30% in p53, and 3% in MutS. The results suggest that the significance of pathways may have system specific factors involved. Furthermore, the possible existence of false positives for contiguous pathways implies that signaling could be occurring via alternate routes including those consistent with the energetic landscape model.
Collapse
Affiliation(s)
- Kelly M. Thayer
- Department of Mathematics and Computer Science, Wesleyan University, Middletown, CT, United States of America
- Program in Molecular Biophysics, Wesleyan University, Middletown, CT, United States of America
- Department of Chemistry, Wesleyan University, Middletown, CT, United States of America
- * E-mail:
| | - Jesse C. Galganov
- Department of Mathematics and Computer Science, Wesleyan University, Middletown, CT, United States of America
- Program in Bioinformatics, Wesleyan University, Middletown, CT, United States of America
| | - Avram J. Stein
- Department of Astronomy, Wesleyan University, Middletown, CT, United States of America
- Department of Earth and Environmental Sciences, Wesleyan University, Middletown, CT, United States of America
| |
Collapse
|
38
|
Mast N, Saadane A, Valencia-Olvera A, Constans J, Maxfield E, Arakawa H, Li Y, Landreth G, Pikuleva IA. Cholesterol-metabolizing enzyme cytochrome P450 46A1 as a pharmacologic target for Alzheimer's disease. Neuropharmacology 2017; 123:465-476. [PMID: 28655608 PMCID: PMC5546235 DOI: 10.1016/j.neuropharm.2017.06.026] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 06/21/2017] [Accepted: 06/23/2017] [Indexed: 12/27/2022]
Abstract
Cytochrome P450 46A1 (CYP46A1 or cholesterol 24-hydroxylase) controls cholesterol elimination from the brain and plays a role in higher order brain functions. Genetically enhanced CYP46A1 expression in mouse models of Alzheimer's disease mitigates the manifestations of this disease. We enhanced CYP46A1 activity pharmacologically by treating 5XFAD mice, a model of rapid amyloidogenesis, with a low dose of the anti-HIV medication efavirenz. Efavirenz was administered from 1 to 9 months of age, and mice were evaluated at specific time points. At one month of age, cholesterol homeostasis was already disturbed in the brain of 5XFAD mice. Nevertheless, efavirenz activated CYP46A1 and mouse cerebral cholesterol turnover during the first four months of administration. This treatment time also reduced amyloid burden and microglia activation in the cortex and subiculum of 5XFAD mice as well as protein levels of amyloid precursor protein and the expression of several genes involved in inflammatory response. However, mouse short-term memory and long-term spatial memory were impaired, whereas learning in the context-dependent fear test was improved. Additional four months of drug administration (a total of eight months of treatment) improved long-term spatial memory in the treated as compared to the untreated mice, further decreased amyloid-β content in 5XFAD brain, and also decreased the mortality rate among male mice. We propose a mechanistic model unifying the observed efavirenz effects. We suggest that CYP46A1 activation by efavirenz could be a new anti-Alzheimer's disease treatment and a tool to study and identify normal and pathological brain processes affected by cholesterol maintenance.
Collapse
Affiliation(s)
- Natalia Mast
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Aicha Saadane
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Ana Valencia-Olvera
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - James Constans
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Erin Maxfield
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Hiroyuki Arakawa
- Behavioral Core, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Young Li
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Gary Landreth
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Irina A Pikuleva
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH 44106, USA.
| |
Collapse
|
39
|
Iwatani-Yoshihara M, Ito M, Ishibashi Y, Oki H, Tanaka T, Morishita D, Ito T, Kimura H, Imaeda Y, Aparicio S, Nakanishi A, Kawamoto T. Discovery and Characterization of a Eukaryotic Initiation Factor 4A-3-Selective Inhibitor That Suppresses Nonsense-Mediated mRNA Decay. ACS Chem Biol 2017; 12:1760-1768. [PMID: 28440616 DOI: 10.1021/acschembio.7b00041] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Eukaryotic initiation factor 4A-3 (eIF4A3) is an Asp-Glu-Ala-Asp (DEAD) box-family adenosine triphosphate (ATP)-dependent RNA helicase. Subtypes eIF4A1 and eIF4A2 are required for translation initiation, but eIF4A3 participates in the exon junction complex (EJC) and functions in RNA metabolism including nonsense-mediated RNA decay (NMD). No small molecules for NMD inhibition via selective inhibition of eIF4A3 have been discovered. Here, we identified allosteric eIF4A3 inhibitors from a high-throughput screening campaign. Chemical optimization of the lead compounds based on ATPase activity yielded compound 2, which exhibited noncompetitive inhibition with ATP or RNA and high selectivity for eIF4A3 over other helicases. The optimized compounds suppressed the helicase activity of eIF4A3 in an ATPase-dependent manner. Hydrogen/deuterium exchange mass spectrometry demonstrated that the deuterium-incorporation pattern of compound 2 overlapped with that of an allosteric pan-eIF4A inhibitor, hippuristanol, suggesting that compound 2 binds to an allosteric region on eIF4A3. We examined NMD activity using a luciferase-based cellular reporter system and a quantitative real-time polymerase chain-reaction-based cellular system to monitor levels of endogenous NMD substrates. NMD suppression by the compounds correlated positively with their ATPase-inhibitory activity. In conclusion, we developed a novel eIF4A3 inhibitor that targets the EJC. The optimized chemical probes represent useful tools for understanding the functions of eIF4A3 in RNA homeostasis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Samuel Aparicio
- BC Cancer Agency, Department of Molecular Oncology, Vancouver, British Columbia V5Z 1L3, Canada
- University of British Columbia, Department of Pathology
and Laboratory Medicine, Vancouver, British Columbia V6T 2B5, Canada
| | | | | |
Collapse
|
40
|
Mast N, Anderson KW, Johnson KM, Phan TTN, Guengerich FP, Pikuleva IA. In vitro cytochrome P450 46A1 (CYP46A1) activation by neuroactive compounds. J Biol Chem 2017. [PMID: 28642370 DOI: 10.1074/jbc.m117.794909] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cytochrome P450 46A1 (CYP46A1, cholesterol 24-hydroxylase) is the enzyme responsible for the majority of cholesterol elimination from the brain. Previously, we found that the anti-HIV drug efavirenz (EFV) can pharmacologically activate CYP46A1 in mice. Herein, we investigated whether CYP46A1 could also be activated by endogenous compounds, including major neurotransmitters. In vitro experiments with purified recombinant CYP46A1 indicated that CYP46A1 is activated by l-glutamate (l-Glu), l-aspartate, γ-aminobutyric acid, and acetylcholine, with l-Glu eliciting the highest increase (3-fold) in CYP46A1-mediated cholesterol 24-hydroxylation. We also found that l-Glu and other activating neurotransmitters bind to the same site on the CYP46A1 surface, which differs from the EFV-binding site. The other principal differences between EFV and l-Glu in CYP46A1 activation include an apparent lack of l-Glu binding to the P450 active site and different pathways of signal transduction from the allosteric site to the active site. EFV and l-Glu similarly increased the CYP46A1 kcat, the rate of the "fast" phase of the enzyme reduction by the redox partner NADPH-cytochrome P450 oxidoreductase, and the amount of P450 reduced. Spectral titrations with cholesterol, in the presence of EFV or l-Glu, suggest that water displacement from the heme iron can be affected in activator-bound CYP46A1. Moreover, EFV and l-Glu synergistically activated CYP46A1. Collectively, our in vitro data, along with those from previous cell culture and in vivo studies by others, suggest that l-Glu-induced CYP46A1 activation is of physiological relevance.
Collapse
Affiliation(s)
- Natalia Mast
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, Ohio 44106
| | - Kyle W Anderson
- Biomolecular Measurement Division, National Institute of Standards and Technology, Gaithersburg, Maryland 20899; Institute for Bioscience and Biotechnology Research, Rockville, Maryland 20850
| | - Kevin M Johnson
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232
| | - Thanh T N Phan
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232
| | - F Peter Guengerich
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232
| | - Irina A Pikuleva
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, Ohio 44106.
| |
Collapse
|
41
|
Abstract
Membrane proteins play a most important part in metabolism, signaling, cell motility, transport, development, and many other biochemical and biophysical processes which constitute fundamentals of life on the molecular level. Detailed understanding of these processes is necessary for the progress of life sciences and biomedical applications. Nanodiscs provide a new and powerful tool for a broad spectrum of biochemical and biophysical studies of membrane proteins and are commonly acknowledged as an optimal membrane mimetic system that provides control over size, composition, and specific functional modifications on the nanometer scale. In this review we attempted to combine a comprehensive list of various applications of nanodisc technology with systematic analysis of the most attractive features of this system and advantages provided by nanodiscs for structural and mechanistic studies of membrane proteins.
Collapse
Affiliation(s)
- Ilia G Denisov
- Department of Biochemistry and Department of Chemistry, University of Illinois , Urbana, Illinois 61801, United States
| | - Stephen G Sligar
- Department of Biochemistry and Department of Chemistry, University of Illinois , Urbana, Illinois 61801, United States
| |
Collapse
|
42
|
Mast N, Anderson KW, Lin JB, Li Y, Turko IV, Tatsuoka C, Bjorkhem I, Pikuleva IA. Cytochrome P450 27A1 Deficiency and Regional Differences in Brain Sterol Metabolism Cause Preferential Cholestanol Accumulation in the Cerebellum. J Biol Chem 2017; 292:4913-4924. [PMID: 28190002 DOI: 10.1074/jbc.m116.774760] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 02/10/2017] [Indexed: 01/04/2023] Open
Abstract
Cytochrome P450 27A1 (CYP27A1 or sterol 27-hydroxylase) is a ubiquitous, multifunctional enzyme catalyzing regio- and stereospecific hydroxylation of different sterols. In humans, complete CYP27A1 deficiency leads to cerebrotendinous xanthomatosis or nodule formation in tendons and brain (preferentially in the cerebellum) rich in cholesterol and cholestanol, the 5α-saturated analog of cholesterol. In Cyp27a1-/- mice, xanthomas are not formed, despite a significant cholestanol increase in the brain and cerebellum. The mechanism behind cholestanol production has been clarified, yet little is known about its metabolism, except that CYP27A1 might metabolize cholestanol. It also is unclear why CYP27A1 deficiency results in preferential cholestanol accumulation in the cerebellum. We hypothesized that cholestanol might be metabolized by CYP46A1, the principal cholesterol 24-hydroxylase in the brain. We quantified sterols along with CYP27A1 and CYP46A1 in mouse models (Cyp27a1-/-, Cyp46a1-/-, Cyp27a1-/-Cyp46a1-/-, and two wild type strains) and human brain specimens. In vitro experiments with purified P450s were conducted as well. We demonstrate that CYP46A1 is involved in cholestanol removal from the brain and that several factors contribute to the preferential increase in cholestanol in the cerebellum arising from CYP27A1 deficiency. These factors include (i) low cerebellar abundance of CYP46A1 and high cerebellar abundance of CYP27A1, the lack of which probably selectively increases the cerebellar cholestanol production; (ii) spatial separation in the cerebellum of cholesterol/cholestanol-metabolizing P450s from a pool of metabolically available cholestanol; and (iii) weak cerebellar regulation of cholesterol biosynthesis. We identified a new physiological role of CYP46A1, an important brain enzyme and cytochrome P450 that could be activated pharmacologically.
Collapse
Affiliation(s)
- Natalia Mast
- From the Departments of Ophthalmology and Visual Sciences and
| | - Kyle W Anderson
- the Biomolecular Measurement Division, National Institute of Standards and Technology, Gaithersburg, Maryland 20899.,the Institute for Bioscience and Biotechnology Research, Rockville, Maryland 20850, and
| | - Joseph B Lin
- From the Departments of Ophthalmology and Visual Sciences and
| | - Yong Li
- From the Departments of Ophthalmology and Visual Sciences and
| | - Illarion V Turko
- the Biomolecular Measurement Division, National Institute of Standards and Technology, Gaithersburg, Maryland 20899.,the Institute for Bioscience and Biotechnology Research, Rockville, Maryland 20850, and
| | - Curtis Tatsuoka
- Neurology, Case Western Reserve University, Cleveland, Ohio 44106
| | - Ingemar Bjorkhem
- the Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institute, 141 86 Huddinge, Sweden
| | | |
Collapse
|
43
|
Guengerich FP. Intersection of the Roles of Cytochrome P450 Enzymes with Xenobiotic and Endogenous Substrates: Relevance to Toxicity and Drug Interactions. Chem Res Toxicol 2017; 30:2-12. [PMID: 27472660 PMCID: PMC5293730 DOI: 10.1021/acs.chemrestox.6b00226] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Today much is known about cytochrome P450 (P450) enzymes and their catalytic specificity, but the range of reactions catalyzed by each still continues to surprise. Historically, P450s had been considered to be involved in either the metabolism of xenobiotics or endogenous chemicals, in the former case playing a generally protective role and in the latter case a defined physiological role. However, the line of demarcation is sometimes blurred. It is difficult to be completely specific in drug design, and some P450s involved in the metabolism of steroids and vitamins can be off-targets. In a number of cases, drugs have been developed that act on some of those P450s as primary targets, e.g., steroid aromatase inhibitors. Several of the P450s involved in the metabolism of endogenous substrates are less specific than once thought and oxidize several related structures. Some of the P450s that primarily oxidize endogenous chemicals have been shown to oxidize xenobiotic chemicals, even in a bioactivation mode.
Collapse
Affiliation(s)
- F Peter Guengerich
- Department of Biochemistry, Vanderbilt University School of Medicine , Nashville, Tennessee 37232-0146, United States
| |
Collapse
|
44
|
Moutinho M, Nunes MJ, Rodrigues E. Cholesterol 24-hydroxylase: Brain cholesterol metabolism and beyond. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:1911-1920. [PMID: 27663182 DOI: 10.1016/j.bbalip.2016.09.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 09/05/2016] [Accepted: 09/16/2016] [Indexed: 01/19/2023]
Abstract
Dysfunctions in brain cholesterol homeostasis have been extensively related to brain disorders. The major elimination pathway of brain cholesterol is its hydroxylation into 24 (S)-hydroxycholesterol by the cholesterol 24-hydroxylase (CYP46A1). Interestingly, there seems to be an association between CYP46A1 and high-order brain functions, in a sense that increased expression of this hydroxylase improves cognition, while a reduction leads to a poor cognitive performance. Moreover, increasing amount of epidemiological, biochemical and molecular evidence, suggests that CYP46A1 has a role in the pathogenesis or progression of neurodegenerative disorders, in which up-regulation of this enzyme is clearly beneficial. However, the mechanisms underlying these effects are poorly understood, which highlights the importance of studies that further explore the role of CYP46A1 in the central nervous system. In this review we summarize the major findings regarding CYP46A1, and highlight the several recently described pathways modulated by this enzyme from a physiological and pathological perspective, which might account for novel therapeutic strategies for neurodegenerative disorders.
Collapse
Affiliation(s)
- Miguel Moutinho
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Maria João Nunes
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Elsa Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
| |
Collapse
|