1
|
Tsoi SC, Barrientos AC, Vicario DS, Phan ML, Pytte CL. Daily high doses of atorvastatin alter neuronal morphology in a juvenile songbird model. PLoS One 2025; 20:e0314690. [PMID: 40294005 PMCID: PMC12036933 DOI: 10.1371/journal.pone.0314690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 11/11/2024] [Indexed: 04/30/2025] Open
Abstract
Statins are highly effective and widely prescribed cholesterol lowering drugs. However, statins cross the blood-brain barrier and decrease neural cholesterol in animal models, raising concern that long-term statin use may impact cholesterol-dependent structures and functions in the brain. Cholesterol is a fundamental component of cell membranes and experimentally decreasing membrane cholesterol has been shown to alter cell morphology in vitro. In addition, brain regions that undergo adult neurogenesis rely on local brain cholesterol for the manufacture of new neuronal membranes. Thus neurogenesis may be particularly vulnerable to long-term statin use. Here we asked whether oral statin treatment impacts neurogenesis in juveniles, either by decreasing numbers of new cells formed or altering the structure of new neurons. The use of statins in children and adolescents has received less attention than in older adults, with few studies on potential unintended effects in young brains. We examined neurons in the juvenile zebra finch songbird in telencephalic regions that function in song perception and memory (caudomedial nidopallium, NCM) and song production (HVC). Birds received either 40 mg/kg of atorvastatin in water or water vehicle once daily for 2-3 months until they reached adulthood. We labeled newborn cells using systemic injections of bromodeoxyuridine (BrdU) and quantified cells double-labeled with antibodies for BrdU and the neuron-specific protein Hu 30-32 days post mitosis. We also quantified a younger cohort of new neurons in the same birds using antibody to the neuronal protein doublecortin (DCX). We then compared numbers of new neurons and soma morphology of BrdU + /Hu+ neurons between statin-treated and control birds. We did not find an effect of statins on the density of newly formed neurons in either brain region, suggesting that statin treatment did not impact neurogenesis or young neuron survival in our paradigm. However, we found that neuronal soma morphology differed significantly between statin-treated and control birds. Somata of BrdU + /Hu+ (30-32 day old) neurons were flatter and had more furrowed contours in statin-treated birds relative to controls. In a larger, heterogeneous cohort of non-birthdated BrdU-/Hu+ neurons, largely born prior to statin treatment, somata were smaller in statin-treated birds than in controls. Our findings indicate that atorvastatin may affect neural cytoarchitecture in both newly formed and mature neurons, perhaps as a consequence of decreased cholesterol availability in the brain.
Collapse
Affiliation(s)
- Shuk C. Tsoi
- CUNY Neuroscience Collaborative, Psychology and Biology Departments, The Graduate Center, City University of New York, New York, New York, United States of America
| | - Alicia C. Barrientos
- CUNY Neuroscience Collaborative, Psychology and Biology Departments, The Graduate Center, City University of New York, New York, New York, United States of America
| | - David S. Vicario
- Department of Psychology, Rutgers, The State University of New Jersey, Piscataway, New Jersey, United States of America
| | - Mimi L. Phan
- Department of Psychology, Rutgers, The State University of New Jersey, Piscataway, New Jersey, United States of America
| | - Carolyn L. Pytte
- CUNY Neuroscience Collaborative, Psychology and Biology Departments, The Graduate Center, City University of New York, New York, New York, United States of America
- Psychology Department, Queens College, City University of New York, Flushing, New York, United States of America
| |
Collapse
|
2
|
Vanherle S, Loix M, Miron VE, Hendriks JJA, Bogie JFJ. Lipid metabolism, remodelling and intercellular transfer in the CNS. Nat Rev Neurosci 2025; 26:214-231. [PMID: 39972160 DOI: 10.1038/s41583-025-00908-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2025] [Indexed: 02/21/2025]
Abstract
Lipid metabolism encompasses the catabolism and anabolism of lipids, and is fundamental for the maintenance of cellular homeostasis, particularly within the lipid-rich CNS. Increasing evidence further underscores the importance of lipid remodelling and transfer within and between glial cells and neurons as key orchestrators of CNS lipid homeostasis. In this Review, we summarize and discuss the complex landscape of processes involved in lipid metabolism, remodelling and intercellular transfer in the CNS. Highlighted are key pathways, including those mediating lipid (and lipid droplet) biogenesis and breakdown, lipid oxidation and phospholipid metabolism, as well as cell-cell lipid transfer mediated via lipoproteins, extracellular vesicles and tunnelling nanotubes. We further explore how the dysregulation of these pathways contributes to the onset and progression of neurodegenerative diseases, and examine the homeostatic and pathogenic impacts of environment, diet and lifestyle on CNS lipid metabolism.
Collapse
Affiliation(s)
- Sam Vanherle
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
- University MS Centre, Hasselt University, Hasselt, Belgium
| | - Melanie Loix
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
- University MS Centre, Hasselt University, Hasselt, Belgium
| | - Veronique E Miron
- Keenan Research Centre for Biomedical Science and Barlo Multiple Sclerosis Centre, St Michael's Hospital, Toronto, Ontario, Canada
- Department of Immunology, The University of Toronto, Toronto, Ontario, Canada
- UK Dementia Research Institute at The University of Edinburgh, Edinburgh, UK
| | - Jerome J A Hendriks
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
- University MS Centre, Hasselt University, Hasselt, Belgium
| | - Jeroen F J Bogie
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Hasselt, Belgium.
- University MS Centre, Hasselt University, Hasselt, Belgium.
| |
Collapse
|
3
|
Duan C, Luo C, Jiang W, Xu H, Chen Y, Xu S, Zhang X, Chen X, He D. Predictive factors and risk model for depression in patients with type 2 diabetes mellitus: a comprehensive analysis of comorbidities and clinical indicators. Front Endocrinol (Lausanne) 2025; 16:1555142. [PMID: 40110545 PMCID: PMC11919683 DOI: 10.3389/fendo.2025.1555142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 02/17/2025] [Indexed: 03/22/2025] Open
Abstract
Objective Depression is highly prevalent among individuals with type 2 diabetes mellitus (T2DM), often compounded by multiple chronic conditions. This study aimed to identify the key factors influencing depression in this population, with a particular focus on the relationship between the Cumulative Illness Rating Scale (CIRS) score and depression, and to evaluate the predictive value of a model incorporating sex, body mass index (BMI), low-density lipoprotein cholesterol (LDL-C), and CIRS score. Methods A total of 308 hospitalized patients with type 2 diabetes from Quzhou Hospital, Wenzhou Medical University were enrolled. Their clinical and biochemical data were collected, alongside assessments of comorbidities and depressive symptoms using the CIRS and Self-Rating Depression Scale (SDS), respectively. LASSO regression with 10-fold cross-validation was used to identify the optimal variables for the predictive model. Multivariate analysis was performed to assess the independent associations between sex, BMI, LDL-C, and CIRS score with depression. The relationship between CIRS scores and depression was further explored across various subgroups. The predictive model's value was assessed through ROC curve analysis. Results Female sex (OR: 2.48, 95% CI: 1.50-4.10, p < 0.001), lower BMI (OR: 0.92, 95% CI: 0.86-0.98, p = 0.015), lower LDL-C (OR: 0.77, 95% CI: 0.61-0.98, p = 0.031), and higher CIRS scores (OR: 1.11, 95% CI: 1.05-1.18, p < 0.001) were independently linked to depression after adjusting for clinical variables. A strong association between CIRS score and depression was observed, particularly in males, patients under 60 years old, those with a disease duration of less than 5 years, and individuals with no history of smoking or alcohol consumption. Additionally, a predictive model incorporating sex, BMI, LDL-C, and CIRS score demonstrated high accuracy in identifying patients at risk for depression. Conclusions Female, lower BMI, lower LDL-C and higher CIRS score were independently associated with depression in patients with type 2 diabetes. The CIRS score appeared to be more effective in predicting depression risk in people who were male, younger, shorter DM duration, no smoking or no drinking. A more comprehensive prediction model could help clinicians identify patients with type 2 diabetes who are at risk for depression.
Collapse
Affiliation(s)
- Chengzheng Duan
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Quzhou, China
| | - Cheng Luo
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Quzhou, China
| | - Weifeng Jiang
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Quzhou, China
| | - Hui Xu
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Quzhou, China
| | - Yexing Chen
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shiyu Xu
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Quzhou, China
| | - Xiaofang Zhang
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Quzhou, China
| | - Xiaoli Chen
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Quzhou, China
| | - Dongjuan He
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Quzhou, China
| |
Collapse
|
4
|
Boccardi V, Mancinetti F, Guazzarini AG, Murasecco I, Melis F, Bastiani P, Scamosci M, Cecchetti R, Mecocci P. Sex-specific associations between serum lipid levels and cognitive performance in older adults: results from a cross-sectional real-world study. Aging Clin Exp Res 2025; 37:62. [PMID: 40021569 PMCID: PMC11870876 DOI: 10.1007/s40520-025-02976-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 02/17/2025] [Indexed: 03/03/2025]
Abstract
AIM Dyslipidemia and cognitive decline are prevalent in older adults, with their incidence increasing with age. However, the relationship between serum lipid levels and cognitive dysfunction in geriatrics remains unclear, potentially influenced by sex differences. METHODS This study evaluated serum lipid levels and cognitive functions in older adults using a large battery of neuropsychological tests. Dementia was staged with the Clinical Dementia Rating (CDR), classifying participants as cognitively healthy (CDR 0), mildly impaired (CDR 0.5), or with dementia (CDR ≥ 1). RESULTS The study involved 1283 participants aged over 65 (466 men, 817 women; mean age 79.79 ± 5.93 years). Women had lower education levels, reduced autonomy in activities of daily living (ADL), but greater independence in instrumental ADL. Additionally, women exhibited lower glucose but higher levels of total cholesterol (TC), high-density lipoprotein (HDL-C), and low-density lipoprotein (LDL-C) compared to men. Subjects with CDR ≥ 1 had significantly poorer cognitive scores than those with CDR 0 or 0.5. No associations were found between lipid levels and cognition in the CDR 0 group. In men with CDR 0, HDL-C positively correlated with ACE-R Fluency. In the CDR 0.5 group, TC and HDL-C were linked to better cognitive performance. For CDR ≥ 1, TC and HDL-C were associated with improved cognition in women but linked to cognitive decline in men. CONCLUSION Elevated late-life cholesterol may protect cognitive function in healthy individuals and those with mild impairment, with a sex-specific impact in dementia, beneficial for women but detrimental for men.
Collapse
Affiliation(s)
- Virginia Boccardi
- Division of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Piazzale Gambuli 1, 06132, Perugia, Italy.
| | - Francesca Mancinetti
- Division of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Piazzale Gambuli 1, 06132, Perugia, Italy
| | - Anna Giulia Guazzarini
- Division of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Piazzale Gambuli 1, 06132, Perugia, Italy
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Ilenia Murasecco
- Division of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Piazzale Gambuli 1, 06132, Perugia, Italy
| | - Francesco Melis
- Division of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Piazzale Gambuli 1, 06132, Perugia, Italy
| | - Patrizia Bastiani
- Division of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Piazzale Gambuli 1, 06132, Perugia, Italy
| | - Michela Scamosci
- Division of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Piazzale Gambuli 1, 06132, Perugia, Italy
| | - Roberta Cecchetti
- Division of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Piazzale Gambuli 1, 06132, Perugia, Italy
| | - Patrizia Mecocci
- Division of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Piazzale Gambuli 1, 06132, Perugia, Italy
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
5
|
Monnot C, Kalomoiri M, MacNicol E, Kim E, Mesquita M, Damberg P, Van Kampen JM, Kay DG, Turkheimer F, Robertson HA, Cash D, Svenningsson P. Early alterations of functional connectivity, regional brain volumes and astrocyte markers in the beta-sitosterol beta-d-glucoside (BSSG) rat model of parkinsonism. Exp Neurol 2025; 385:115118. [PMID: 39716587 DOI: 10.1016/j.expneurol.2024.115118] [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: 08/12/2024] [Revised: 12/01/2024] [Accepted: 12/15/2024] [Indexed: 12/25/2024]
Abstract
The β-sitosterol-β-ᴅ-glucoside (BSSG) rat model of experimental parkinsonism develops pathological behaviour and motor changes that progress over time. The purpose of this study was to identify early changes in structure and function of the brain of rats treated with BSSG using both structural and resting-state functional MRI. BSSG and non-BSSG rats were fed five days a week for sixteen weeks, then underwent in vivo MRI scans and an assessment of motor performance 2 and 8 weeks later (18 and week 24 from BSSG). Groups of rats were killed at weeks 19 and 25, then imaged again with MR ex vivo, and immunostained for tyrosine hydroxylase (TH). Since BSSG may interfere with cholesterol metabolism in astrocytes, we also studied potential target engagement and measured levels of astrocyte markers GFAP and S100b. At both studied timepoints, functional connectivity (FC) between brain areas with intermediate connectivity was decreased, but brain volumes increased in the BSSG-treated rats. At week 18/19, fine movements were normal, whereas TH and GFAP were decreased in the striatum, but not in the substantia nigra. At week 24/25, fine movements were impaired, and TH was decreased both in the striatum and the substantia nigra and S100b was increased in the substantia nigra. Astrogliosis may contribute to the increased brain volume found after BSSG exposure. Using the BSSG model of parkinsonism, the results demonstrate early functional and structural alterations in MRI imaging that occur before the appearance of detectable motor symptoms.
Collapse
Affiliation(s)
- C Monnot
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - M Kalomoiri
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - E MacNicol
- Department of Neuroimaging, Institute of Psychology, Psychiatry and Neuroscience, King's College London, London, UK
| | - E Kim
- Department of Neuroimaging, Institute of Psychology, Psychiatry and Neuroscience, King's College London, London, UK
| | - M Mesquita
- Department of Neuroimaging, Institute of Psychology, Psychiatry and Neuroscience, King's College London, London, UK
| | - P Damberg
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - J M Van Kampen
- Neurodyn Life Sciences Inc., Charlottetown, Prince Edward Island, Canada
| | - D G Kay
- Neurodyn Life Sciences Inc., Charlottetown, Prince Edward Island, Canada
| | - F Turkheimer
- Department of Neuroimaging, Institute of Psychology, Psychiatry and Neuroscience, King's College London, London, UK
| | - H A Robertson
- Neurodyn Life Sciences Inc., Charlottetown, Prince Edward Island, Canada
| | - D Cash
- Department of Neuroimaging, Institute of Psychology, Psychiatry and Neuroscience, King's College London, London, UK.
| | - P Svenningsson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden; Department of Basic and Clinical Neuroscience, King's College London, London, UK.
| |
Collapse
|
6
|
Savulescu-Fiedler I, Dorobantu-Lungu LR, Dragosloveanu S, Benea SN, Dragosloveanu CDM, Caruntu A, Scheau AE, Caruntu C, Scheau C. The Cross-Talk Between the Peripheral and Brain Cholesterol Metabolisms. Curr Issues Mol Biol 2025; 47:115. [PMID: 39996836 PMCID: PMC11853762 DOI: 10.3390/cimb47020115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 01/30/2025] [Accepted: 02/07/2025] [Indexed: 02/26/2025] Open
Abstract
Cholesterol is an essential element for the development and normal function of the central nervous system. While peripheral cholesterol is influenced by liver metabolism and diet, brain cholesterol metabolism takes place in an isolated system due to the impermeability of the blood-brain barrier (BBB). However, cross-talk occurs between the brain and periphery, specifically through metabolites such as oxysterols that play key roles in regulating cholesterol balance. Several neurodegenerative conditions such as Alzheimer's disease or Parkinson's disease are considered to be affected by the loss of this balance. Also, the treatment of hypercholesterolemia needs to consider these discrete interferences between brain and peripheral cholesterol and the possible implications of each therapeutic approach. This is particularly important because of 27-hydroxycholesterol and 24-hydroxycholesterol, which can cross the BBB and are involved in cholesterol metabolism. This paper examines the metabolic pathways of cholesterol metabolism in the brain and periphery and focuses on the complex cross-talk between these metabolisms. Also, we emphasize the regulatory role of the BBB and the need for an integrated approach to cholesterol management.
Collapse
Affiliation(s)
- Ilinca Savulescu-Fiedler
- Department of Internal Medicine, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Internal Medicine and Cardiology, Coltea Clinical Hospital, 030167 Bucharest, Romania
| | - Luiza-Roxana Dorobantu-Lungu
- Department of Cardiology, Emergency Institute for Cardiovascular Diseases “C.C. Iliescu”, 022328 Bucharest, Romania
| | - Serban Dragosloveanu
- Department of Orthopaedics, “Foisor” Clinical Hospital of Orthopaedics, Traumatology and Osteoarticular TB, 021382 Bucharest, Romania
- Department of Orthopaedics and Traumatology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Serban Nicolae Benea
- Department of Infectious Diseases, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Departament of Infectious Diseases, National Institute for Infectious Diseases “Prof. Dr. Matei Balș”, 021105 Bucharest, Romania
| | - Christiana Diana Maria Dragosloveanu
- Department of Ophthalmology, Faculty of Dentistry, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Ophthalmology, Clinical Hospital for Ophthalmological Emergencies, 010464 Bucharest, Romania
| | - Ana Caruntu
- Department of Oral and Maxillofacial Surgery, “Carol Davila” Central Military Emergency Hospital, 010825 Bucharest, Romania
- Department of Oral and Maxillofacial Surgery, Faculty of Dental Medicine, “Titu Maiorescu” University, 031593 Bucharest, Romania
| | - Andreea-Elena Scheau
- Department of Radiology and Medical Imaging, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Constantin Caruntu
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Dermatology, “Prof. N.C. Paulescu” National Institute of Diabetes, Nutrition and Metabolic Diseases, 011233 Bucharest, Romania
| | - Cristian Scheau
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Radiology and Medical Imaging, “Foisor” Clinical Hospital of Orthopaedics, Traumatology and Osteoarticular TB, 021382 Bucharest, Romania
| |
Collapse
|
7
|
Gence L, Morello E, Rastegar S, Apalama ML, Meilhac O, Bascands JL, Diotel N. Gene expression patterns of the LDL receptor and its inhibitor Pcsk9 in the adult zebrafish brain suggest a possible role in neurogenesis. Eur J Neurosci 2025; 61:e16586. [PMID: 39551948 DOI: 10.1111/ejn.16586] [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: 11/28/2023] [Revised: 09/18/2024] [Accepted: 10/16/2024] [Indexed: 11/19/2024]
Abstract
The low-density lipoprotein receptor (LDLr) is the first member of a closely related transmembrane protein family. It is known for its involvement in various physiological processes, mainly in the regulation of lipid metabolism, especially in the brains of mammals and zebrafish. In zebrafish, two ldlr genes (ldlra and b) have been identified and their distribution in the brain is not well documented. Recently, the roles of ldlr and its inhibitor pcsk9 in regenerative process after telencephalic brain injury have been discussed. In this study, we explored the expression patterns of these genes during zebrafish development. We found that ldlra expression was detected at the end of the pharyngula period (48 hpf) and increased during the larval stage. Conversely, ldlrb expression was observed from zygotic to larval stages. Using techniques like in situ hybridization and taking advantage of transgenic fish, we demonstrated the widespread distribution of ldlra, ldlrb and pcsk9 in the brain of adult zebrafish. Specifically, these genes were expressed in neurons and neural stem cells and also at lower levels in endothelial cells. As expected, intraperitoneal injection of fluorescent-labelled LDLs resulted in their uptake by cerebral endothelial cells in a homeostatic context, whereas they diffused within the brain parenchyma after telencephalic injury. However, after intracerebroventricular injections into animals, LDL particles were not taken up by neural stem cells. In conclusion, our results provide additional evidence for LDLr expression in the brain of adult zebrafish. These results raise the question of the role of LDLr in the cholesterol/lipid imbalance in cerebral complications.
Collapse
Affiliation(s)
- Laura Gence
- Université de La Réunion, INSERM, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Saint-Pierre, La Réunion, France
| | - Elena Morello
- Université de La Réunion, INSERM, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Saint-Pierre, La Réunion, France
- CHU de La Réunion, Saint-Pierre, La Réunion, France
| | - Sepand Rastegar
- Institute of Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Marie Laurine Apalama
- Université de La Réunion, INSERM, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Saint-Pierre, La Réunion, France
| | - Olivier Meilhac
- Université de La Réunion, INSERM, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Saint-Pierre, La Réunion, France
- CHU de La Réunion, Saint-Pierre, La Réunion, France
| | - Jean-Loup Bascands
- Université de La Réunion, INSERM, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Saint-Pierre, La Réunion, France
| | - Nicolas Diotel
- Université de La Réunion, INSERM, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), Saint-Pierre, La Réunion, France
| |
Collapse
|
8
|
Borràs C, Canyelles M, Santos D, Rotllan N, Núñez E, Vázquez J, Maspoch D, Cano-Sarabia M, Carmona-Iragui M, Sirisi S, Lleó A, Fortea J, Alcolea D, Blanco-Vaca F, Escolà-Gil JC, Tondo M. Impaired Cerebrospinal Fluid Lipoprotein-Mediated Cholesterol Delivery to Neurons in Alzheimer's Disease. RESEARCH SQUARE 2024:rs.3.rs-5682870. [PMID: 39764088 PMCID: PMC11703344 DOI: 10.21203/rs.3.rs-5682870/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2025]
Abstract
In the central nervous system, apolipoprotein (APO) E-containing high-density lipoprotein (HDL)-like particles mediate the transport of glial-derived cholesterol to neurons, which is essential for neuronal membrane remodeling and maintenance of the myelin sheath. Despite this, the role of HDL-like cholesterol trafficking on Alzheimer's disease (AD) pathogenesis remains poorly understood. We aimed to examine cholesterol transport via HDL-like particles in cerebrospinal fluid (CSF) of AD patients compared to control individuals. Additionally, we explored the ability of reconstituted HDL containing different APOE isoforms to regulate cholesterol transport. We evaluated the capacity of CSF HDL-like particles to facilitate radiolabeled unesterified cholesterol efflux from A172 human glioblastoma astrocytes and to deliver cholesterol to SH-SY5Y human neuronal cells. The HDL-like proteome in the AD and control groups was analyzed by liquid chromatography-mass spectrometry (LC-MS/MS). Reconstituted HDL nanoparticles were prepared by combining phospholipids and cholesterol with human APOE3 or APOE4, followed by radiolabeling with unesterified cholesterol. Our results showed that cholesterol efflux from astrocytes to CSF were similar between AD patients and controls, both under baseline conditions and after activation of ATP-binding cassette transporters A1 and G1. However, CSF HDL-like particle-mediated neuronal cholesterol uptake was significantly reduced in the AD group. LC-MS/MS analysis identified 775 proteins associated with HDL-like particles in both groups, with no major alterations in proteins linked to cholesterol metabolism. However, 27 proteins involved in non-cholesterol-related processes were differentially expressed. Notably, synthetic reconstituted HDL particles containing APOE4 exhibited reduced capacity to deliver cholesterol to neurons compared to those with APOE3. These findings indicate that CSF HDL-like particles from patients with AD demonstrate impaired cholesterol delivery to neurons. Our study highlights APOE4 as a critical contributor to abnormal neuronal cholesterol uptake in AD pathophysiology.
Collapse
Affiliation(s)
| | | | - David Santos
- Centro de Investigación Biomédica en Red Diabetes y Enfermedades Metabólicas Asociadas
| | | | - Estefanía Núñez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III
| | - Jesús Vázquez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III
| | - Daniel Maspoch
- Catalan Institute of Nanoscience and Nanotechnology (ICN2)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Lorincz B, Vrablik M, Murali R, Havrdova EK, Horakova D, Krasensky J, Vaneckova M, Uher T. Lipid and brain volumetric measures in multiple sclerosis patients: findings from a large observational study. Acta Neurol Belg 2024; 124:1981-1988. [PMID: 39455532 PMCID: PMC11614926 DOI: 10.1007/s13760-024-02676-w] [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: 01/05/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024]
Abstract
OBJECTIVES This study aimed to investigate relationships between cholesterol profile, brain volumetric MRI, and clinical measures in a large observational cohort of multiple sclerosis (MS) patients. MATERIALS AND METHODS We included 1.505 patients with 4.966 time points including complete lipid, clinical, and imaging data. The time among lipid, brain MRI and clinical measures was under 90 days. Cross-sectional statistical analysis at baseline was performed using an adjusted linear regression and analysis of longitudinal lipid and MRI measures data was performed using adjusted linear mixed models. RESULTS We found associations between higher high-density lipoprotein cholesterol (HDL-C) and lower brain parenchymal fraction (BPF) at cross-sectional analysis at baseline (B = -0.43, CI 95%: -0.73, -0.12, p = 0.005), as well as in longitudinal analysis over follow-up (B = -0.32 ± 0.072, χ2 = 36.6; p = < 0.001). Higher HDL-C was also associated with higher T2-lesion volume in longitudinal analysis (B = 0.11 ± 0.023; χ2 = 23.04; p = < 0.001). We observed a weak negative association between low-density lipoprotein cholesterol (LDL-C) levels and BPF at baseline (B = -0.26, CI 95%: -0.4, -0.11, p = < 0.001) as well as in longitudinal analysis (B = -0.06 ± 0.03, χ2 = 4.46; p = 0.03). T2-LV did not show an association with LDL-C. We did not find any association between lipid measures and disability. The effect of lipid levels on MRI measures and disability was minimal (Cohen f2 < 0.02). CONCLUSIONS Our results contradict the previously described exclusively positive effect of HDL-C on brain atrophy in patients with MS. Higher LDL-C was weakly associated with higher brain atrophy but not with higher lesion burden.
Collapse
Affiliation(s)
- Balazs Lorincz
- Department of Neurology, Center of Clinical Neuroscience, First Faculty of Medicine, Charles University in Prague and General University Hospital, Prague, Czech Republic
| | - Michal Vrablik
- Third Department of Internal Medicine, Department of Endocrinology and Metabolism, First Faculty of Medicine, Charles University in Prague and General University Hospital, Prague, Czech Republic
| | - Ramanathan Murali
- Department of Pharmaceutical Sciences and Neurology, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Eva Kubala Havrdova
- Department of Neurology, Center of Clinical Neuroscience, First Faculty of Medicine, Charles University in Prague and General University Hospital, Prague, Czech Republic
| | - Dana Horakova
- Department of Neurology, Center of Clinical Neuroscience, First Faculty of Medicine, Charles University in Prague and General University Hospital, Prague, Czech Republic
| | - Jan Krasensky
- Department of Radiology, First Faculty of Medicine, Charles University in Prague and General University Hospital, Prague, Czech Republic
| | - Manuela Vaneckova
- Department of Radiology, First Faculty of Medicine, Charles University in Prague and General University Hospital, Prague, Czech Republic
| | - Tomas Uher
- Department of Neurology, Center of Clinical Neuroscience, First Faculty of Medicine, Charles University in Prague and General University Hospital, Prague, Czech Republic.
| |
Collapse
|
10
|
Mohammedsaeed W. Exploring the interplay between DHCR7, vitamin D deficiency, and type 2 diabetes mellitus (T2DM): a systematic review. Mol Biol Rep 2024; 51:1123. [PMID: 39503960 DOI: 10.1007/s11033-024-10072-z] [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: 06/14/2024] [Accepted: 10/28/2024] [Indexed: 11/20/2024]
Abstract
Type 2 diabetes mellitus (T2DM) is a growing global health concern. The pathogenesis of T2DM is multifactorial and intricate, involving a complex interplay of genetic predisposition, environmental factors, and molecular interactions. Vitamin D (circulating 25-hydroxyvitamin D concentration) regulates factors crucial for T2DM, including insulin secretion, sensitivity, and inflammation. Thus, vitamin D deficiency has been linked to poor health outcomes in T2DM patients. The cholesterol-synthesizing enzyme 7-dehydrocholesterol reductase (DHCR7) represents a critical regulatory switch between cholesterol and vitamin D3 synthesis. Recent findings suggest that the enzyme DHCR7 may indicate T2DM glycolipid metabolic disorder and is associated with deficient circulating vitamin D (circulating 25-hydroxyvitamin D concentration) status. In this PRISMA-guided systematic review, articles were sourced from two databases, namely, PubMed and Cochrane Library, to evaluate the impact of vitamin D deficiency in patients with T2DM and to explore the emerging role of DHCR7 in T2DM pathogenesis. Our findings strongly indicate a positive correlation between deficient vitamin D status and poor health outcomes in T2DM patients. Finally, this systematic review presents a novel perspective on T2DM development, focusing on the interplay between T2DM-associated hyperglycemia, expression of DHCR7, and abrogation of vitamin D synthesis.
Collapse
Affiliation(s)
- Walaa Mohammedsaeed
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Science, Taibah University, 344, Postal Code 3000, Al-Madinah, Saudi Arabia.
| |
Collapse
|
11
|
Gray SM, Dai J, Smith AC, Beckley JT, Rahmati N, Lewis MC, Quirk MC. Changes in 24(S)-Hydroxycholesterol Are Associated with Cognitive Performance in Early Huntington's Disease: Data from the TRACK and ENROLL HD Cohorts. J Huntingtons Dis 2024; 13:449-465. [PMID: 39269850 DOI: 10.3233/jhd-240030] [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] [Indexed: 09/15/2024]
Abstract
BACKGROUND There is evidence for dysregulated cholesterol homeostasis in Huntington's disease (HD). The brain-specific cholesterol metabolite 24(S)-hydroxycholesterol (24(S)-OHC) is decreased in manifest HD. 24(S)-OHC is an endogenous positive allosteric modulator (PAM) of the N-methyl-D-aspartate (NMDA) receptor, suggesting lower 24(S)-OHC may contribute to NMDA receptor hypofunction in HD. We hypothesized changes in 24(S)-OHC would be associated with cognitive impairment in early HD. OBJECTIVE To determine the interactions between oxysterols (24(S)-OHC, 25-OHC, and 27-OHC) at the NMDA receptor, the plasma levels of these oxysterols, and how these levels relate to cognitive performance. METHODS An in vitro competition assay was used to evaluate interactions at the NMDA receptor, liquid chromatography coupled tandem mass spectrometry (LC-MS/MS) was used to measure plasma 24(S)-OHC, 25-OHC, and 27-OHC levels, and correlation analyses investigated their relationship to performance on cognitive endpoints in TRACK and ENROLL-HD (NCT01574053). RESULTS In vitro, 25-OHC and 27-OHC attenuated the PAM activity of 24(S)-OHC on the NMDA receptor. Lower plasma 24(S)-OHC levels and 24(S)/25-OHC ratios were detected in participants with early HD. Moderate and consistent associations were detected between plasma 24(S)/25-OHC ratio and performance on Stroop color naming, symbol digit modality, Trails A/B, and emotion recognition. Little association was observed between the ratio and psychiatric or motor endpoints, suggesting specificity for the relationship to cognitive performance. CONCLUSIONS Our findings support growing evidence for dysregulated CNS cholesterol homeostasis in HD, demonstrate a relationship between changes in oxysterols and cognitive performance in HD, and propose that NMDA receptor hypofunction may contribute to cognitive impairment in HD.
Collapse
Affiliation(s)
| | - Jing Dai
- Sage Therapeutics Inc, Cambridge, MA, USA
| | | | | | | | | | | |
Collapse
|
12
|
Aumont-Rodrigue G, Picard C, Labonté A, Poirier J. Apolipoprotein B gene expression and regulation in relation to Alzheimer's disease pathophysiology. J Lipid Res 2024; 65:100667. [PMID: 39395793 PMCID: PMC11602985 DOI: 10.1016/j.jlr.2024.100667] [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: 04/23/2024] [Revised: 10/04/2024] [Accepted: 10/07/2024] [Indexed: 10/14/2024] Open
Abstract
Apolipoprotein B (APOB), a receptor-binding protein present in cholesterol-rich lipoproteins, has been implicated in Alzheimer's disease (AD). High levels of APOB-containing low-density lipoproteins (LDL) are linked to the pathogenesis of both early-onset familial and late-onset sporadic AD. Rare coding mutations in the APOB gene are associated with familial AD, suggesting a role for APOB-bound lipoproteins in the central nervous system. This research explores APOB gene regulation across the AD spectrum using four cohorts: BRAINEAC (elderly control brains), DBCBB (controls, AD brains), ROSMAP (controls, MCI, AD brains), and ADNI (control, MCI, AD clinical subjects). APOB protein levels, measured via mass spectrometry and ELISA, positively correlated with AD pathology indices and cognition, while APOB mRNA levels showed negative correlations. Brain APOB protein levels are also correlated with cortical Aβ levels. A common coding variant in the APOB gene locus affected its expression but didn't impact AD risk or brain cholesterol concentrations, except for 24-S-hydroxycholesterol. Polymorphisms in the CYP27A1 gene, notably rs4674344, were associated with APOB protein levels. A negative correlation was observed between brain APOB gene expression and AD biomarker levels. CSF APOB correlated with Tau pathology in presymptomatic subjects, while cortical APOB was strongly associated with cortical Aβ deposition in late-stage AD. The study discusses the potential link between blood-brain barrier dysfunction and AD symptoms in relation to APOB neurobiology. Overall, APOB's involvement in lipoprotein metabolism appears to influence AD pathology across different stages of the disease.
Collapse
Affiliation(s)
- Gabriel Aumont-Rodrigue
- Douglas Mental Health University Institute, Neurosciences divison, Montréal, Québec, Canada; Centre for the Studies on Prevention of Alzheimer's Disease, Montréal, Québec, Canada; McGill University, Psychiatry department, Montréal, Québec, Canada
| | - Cynthia Picard
- Douglas Mental Health University Institute, Neurosciences divison, Montréal, Québec, Canada; Centre for the Studies on Prevention of Alzheimer's Disease, Montréal, Québec, Canada
| | - Anne Labonté
- Douglas Mental Health University Institute, Neurosciences divison, Montréal, Québec, Canada; Centre for the Studies on Prevention of Alzheimer's Disease, Montréal, Québec, Canada
| | - Judes Poirier
- Douglas Mental Health University Institute, Neurosciences divison, Montréal, Québec, Canada; Centre for the Studies on Prevention of Alzheimer's Disease, Montréal, Québec, Canada; McGill University, Psychiatry department, Montréal, Québec, Canada.
| |
Collapse
|
13
|
Cao Y, Zhao LW, Chen ZX, Li SH. New insights in lipid metabolism: potential therapeutic targets for the treatment of Alzheimer's disease. Front Neurosci 2024; 18:1430465. [PMID: 39323915 PMCID: PMC11422391 DOI: 10.3389/fnins.2024.1430465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 08/14/2024] [Indexed: 09/27/2024] Open
Abstract
Alzheimer's disease (AD) is increasingly recognized as being intertwined with the dysregulation of lipid metabolism. Lipids are a significant class of nutrients vital to all organisms, playing crucial roles in cellular structure, energy storage, and signaling. Alterations in the levels of various lipids in AD brains and dysregulation of lipid pathways and transportation have been implicated in AD pathogenesis. Clinically, evidence for a high-fat diet firmly links disrupted lipid metabolism to the pathogenesis and progression of AD, although contradictory findings warrant further exploration. In view of the significance of various lipids in brain physiology, the discovery of complex and diverse mechanisms that connect lipid metabolism with AD-related pathophysiology will bring new hope for patients with AD, underscoring the importance of lipid metabolism in AD pathophysiology, and promising targets for therapeutic intervention. Specifically, cholesterol, sphingolipids, and fatty acids have been shown to influence amyloid-beta (Aβ) accumulation and tau hyperphosphorylation, which are hallmarks of AD pathology. Recent studies have highlighted the potential therapeutic targets within lipid metabolism, such as enhancing apolipoprotein E lipidation, activating liver X receptors and retinoid X receptors, and modulating peroxisome proliferator-activated receptors. Ongoing clinical trials are investigating the efficacy of these strategies, including the use of ketogenic diets, statin therapy, and novel compounds like NE3107. The implications of these findings suggest that targeting lipid metabolism could offer new avenues for the treatment and management of AD. By concentrating on alterations in lipid metabolism within the central nervous system and their contribution to AD development, this review aims to shed light on novel research directions and treatment approaches for combating AD, offering hope for the development of more effective management strategies.
Collapse
Affiliation(s)
- Yuan Cao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, Zhengzhou, China
- Clinical Systems Biology Laboratories, Translation Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Lin-Wei Zhao
- Department of Cardiology, People’s Hospital of Zhengzhou University, Henan Provincial People’s Hospital, Zhengzhou University Central China Fuwai Hospital, Zhengzhou, China
| | - Zi-Xin Chen
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, Zhengzhou, China
- Clinical Systems Biology Laboratories, Translation Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Shao-Hua Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, Zhengzhou, China
- Clinical Systems Biology Laboratories, Translation Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
14
|
Koppula S, Wankhede NL, Sammeta SS, Shende PV, Pawar RS, Chimthanawala N, Umare MD, Taksande BG, Upaganlawar AB, Umekar MJ, Kopalli SR, Kale MB. Modulation of cholesterol metabolism with Phytoremedies in Alzheimer's disease: A comprehensive review. Ageing Res Rev 2024; 99:102389. [PMID: 38906182 DOI: 10.1016/j.arr.2024.102389] [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: 04/20/2024] [Revised: 06/18/2024] [Accepted: 06/18/2024] [Indexed: 06/23/2024]
Abstract
Alzheimer's disease (AD) is a complex neurological ailment that causes cognitive decline and memory loss. Cholesterol metabolism dysregulation has emerged as a crucial element in AD pathogenesis, contributing to the formation of amyloid-beta (Aβ) plaques and tau tangles, the disease's hallmark neuropathological characteristics. Thus, targeting cholesterol metabolism has gained attention as a potential therapeutic method for Alzheimer's disease. Phytoremedies, which are generated from plants and herbs, have shown promise as an attainable therapeutic option for Alzheimer's disease. These remedies contain bioactive compounds like phytochemicals, flavonoids, and polyphenols, which have demonstrated potential in modulating cholesterol metabolism and related pathways. This comprehensive review explores the modulation of cholesterol metabolism by phytoremedies in AD. It delves into the role of cholesterol in brain function, highlighting disruptions observed in AD. Additionally, it examines the underlying molecular mechanisms of cholesterol-related pathology in AD. The review emphasizes the significance of phytoremedies as a potential therapeutic intervention for AD. It discusses the drawbacks of current treatments and the need for alternative strategies addressing cholesterol dysregulation and its consequences. Through an in-depth analysis of specific phytoremedies, the review presents compelling evidence of their potential benefits. Molecular mechanisms underlying phytoremedy effects on cholesterol metabolism are examined, including regulation of cholesterol-related pathways, interactions with Aβ pathology, influence on tau pathology, and anti-inflammatory effects. The review also highlights challenges and future perspectives, emphasizing standardization, clinical evidence, and personalized medicine approaches to maximize therapeutic potential in AD treatment. Overall, phytoremedies offer promise as a potential avenue for AD management, but further research and collaboration are necessary to fully explore their efficacy, safety, and mechanisms of action.
Collapse
Affiliation(s)
- Sushruta Koppula
- College of Biomedical and Health Sciences, Konkuk University, Chungju-Si, Chungcheongbuk Do 27478, Republic of Korea.
| | - Nitu L Wankhede
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India.
| | - Shivkumar S Sammeta
- National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana 500037, India.
| | - Prajwali V Shende
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India.
| | - Rupali S Pawar
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India.
| | | | - Mohit D Umare
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India.
| | - Brijesh G Taksande
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India.
| | - Aman B Upaganlawar
- SNJB's Shriman Sureshdada Jain College of Pharmacy, Neminagar, Chandwad, Nashik, Maharashtra, India.
| | - Milind J Umekar
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India.
| | - Spandana Rajendra Kopalli
- Department of Bioscience and Biotechnology, Sejong University, Gwangjin-gu, Seoul 05006, Republic of Korea.
| | - Mayur B Kale
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India.
| |
Collapse
|
15
|
Lu F, Li E, Yang X. Proprotein convertase subtilisin/kexin type 9 deficiency in extrahepatic tissues: emerging considerations. Front Pharmacol 2024; 15:1413123. [PMID: 39139638 PMCID: PMC11319175 DOI: 10.3389/fphar.2024.1413123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 07/08/2024] [Indexed: 08/15/2024] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) is primarily secreted by hepatocytes. PCSK9 is critical in liver low-density lipoprotein receptors (LDLRs) metabolism. In addition to its hepatocellular presence, PCSK9 has also been detected in cardiac, cerebral, islet, renal, adipose, and other tissues. Once perceived primarily as a "harmful factor," PCSK9 has been a focal point for the targeted inhibition of both systemic circulation and localized tissues to treat diseases. However, PCSK9 also contributes to the maintenance of normal physiological functions in numerous extrahepatic tissues, encompassing both LDLR-dependent and -independent pathways. Consequently, PCSK9 deficiency may harm extrahepatic tissues in close association with several pathophysiological processes, such as lipid accumulation, mitochondrial impairment, insulin resistance, and abnormal neural differentiation. This review encapsulates the beneficial effects of PCSK9 on the physiological processes and potential disorders arising from PCSK9 deficiency in extrahepatic tissues. This review also provides a comprehensive analysis of the disparities between experimental and clinical research findings regarding the potential harm associated with PCSK9 deficiency. The aim is to improve the current understanding of the diverse effects of PCSK9 inhibition.
Collapse
Affiliation(s)
- Fengyuan Lu
- The Second Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - En Li
- The Second Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Xiaoyu Yang
- The Second Affiliated Hospital, Zhengzhou University, Zhengzhou, China
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
16
|
Popova EY, Kawasawa YI, Leung M, Barnstable CJ. Temporal changes in mouse hippocampus transcriptome after pilocarpine-induced seizures. Front Neurosci 2024; 18:1384805. [PMID: 39040630 PMCID: PMC11260795 DOI: 10.3389/fnins.2024.1384805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 06/07/2024] [Indexed: 07/24/2024] Open
Abstract
Introduction Status epilepticus (SE) is a seizure lasting more than 5 min that can have lethal consequences or lead to various neurological disorders, including epilepsy. Using a pilocarpine-induced SE model in mice we investigated temporal changes in the hippocampal transcriptome. Methods We performed mRNA-seq and microRNA-seq analyses at various times after drug treatment. Results At 1 h after the start of seizures, hippocampal cells upregulated transcription of immediate early genes and genes involved in the IGF-1, ERK/MAPK and RNA-PolII/transcription pathways. At 8 h, we observed changes in the expression of genes associated with oxidative stress, overall transcription downregulation, particularly for genes related to mitochondrial structure and function, initiation of a stress response through regulation of ribosome and translation/EIF2 signaling, and upregulation of an inflammatory response. During the middle of the latent period, 36 h, we identified upregulation of membrane components, cholesterol synthesis enzymes, channels, and extracellular matrix (ECM), as well as an increased inflammatory response. At the end of the latent period, 120 h, most changes in expression were in genes involved in ion transport, membrane channels, and synapses. Notably, we also elucidated the involvement of novel pathways, such as cholesterol biosynthesis pathways, iron/BMP/ferroptosis pathways, and circadian rhythms signaling in SE and epileptogenesis. Discussion These temporal changes in metabolic reactions indicate an immediate response to injury followed by recovery and regeneration. CREB was identified as the main upstream regulator. Overall, our data provide new insights into molecular functions and cellular processes involved at different stages of seizures and offer potential avenues for effective therapeutic strategies.
Collapse
Affiliation(s)
- Evgenya Y. Popova
- Department of Neural and Behavioral Sciences, Penn State University College of Medicine, Hershey, PA, United States
- Penn State Hershey Eye Center, Hershey, PA, United States
| | - Yuka Imamura Kawasawa
- Department of Pharmacology, Penn State University College of Medicine, Hershey, PA, United States
- Center for Cancer Genomics and Precision Oncology, Wake Forest Baptist Comprehensive Cancer Center, Winston Salem, NC, United States
| | - Ming Leung
- Center for Cancer Genomics and Precision Oncology, Wake Forest Baptist Comprehensive Cancer Center, Winston Salem, NC, United States
| | - Colin J. Barnstable
- Department of Neural and Behavioral Sciences, Penn State University College of Medicine, Hershey, PA, United States
- Penn State Hershey Eye Center, Hershey, PA, United States
| |
Collapse
|
17
|
Magdaleno Herrero R, Murillo-García N, Yorca-Ruiz Á, Neergaard K, Crespo-Facorro B, Ayesa-Arriola R. Biomarkers as proxies for cognitive reserve: The role of high density lipoprotein cholesterol in first episode of psychosis. SPANISH JOURNAL OF PSYCHIATRY AND MENTAL HEALTH 2024; 17:146-153. [PMID: 37852878 DOI: 10.1016/j.rpsm.2023.03.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 01/19/2023] [Accepted: 03/06/2023] [Indexed: 10/20/2023]
Abstract
INTRODUCTION The proxies used to compose cognitive reserve (CR) for patients of a first episode of psychosis (FEP) have varied in the literature. The development of FEP is linked to peripheral pathways of the central nervous system, yet despite this knowledge, no research has considered the introduction of biomarkers as proxies for CR. Meanwhile, schizophrenia has been linked to the metabolic system, indicating that alterations in the levels of biological parameters, in particular high-density lipoproteins (HDL), cause worse global functioning and cognitive impairment. For these reasons, the present study aimed to create a quantifiable and objective CR index that adjusts for the multifactorial nature of FEP. MATERIALS AND METHODS We included 668 FEP patients and 217 healthy controls. Participants were assessed for sociodemographic information, years of education, employment status, premorbid IQ and biological parameters: waist circumference, hypertension, and levels of HDL, triglycerides, and glucose. RESULTS The findings suggest that the years of education proxy showed correlational and higher relationship with HDL levels for both FEP patients (r=0.23, b=0.185) and controls (r=0.31, b=0.342). We found that the CR index composed of years of education and HDL levels showed a higher explanatory power for the phenomenon than the classical CR index composed of years of education, employment status and premorbid IQ. CONCLUSIONS This article proposes an objective and quantifiable method to measure CR that is more the multifactorial nature of FEP.
Collapse
Affiliation(s)
- Rebeca Magdaleno Herrero
- Marqués de Valdecilla University Hospital, IDIVAL, School of Medicine, University of Cantabria, Santander, Spain; Doctoral School University of Cantabria (EDUC), Santander, Spain
| | - Nancy Murillo-García
- Marqués de Valdecilla University Hospital, IDIVAL, School of Medicine, University of Cantabria, Santander, Spain; Doctoral School University of Cantabria (EDUC), Santander, Spain
| | - Ángel Yorca-Ruiz
- Marqués de Valdecilla University Hospital, IDIVAL, School of Medicine, University of Cantabria, Santander, Spain; Doctoral School University of Cantabria (EDUC), Santander, Spain
| | - Karl Neergaard
- Marqués de Valdecilla University Hospital, IDIVAL, School of Medicine, University of Cantabria, Santander, Spain
| | - Benedicto Crespo-Facorro
- Hospital Universitario Virgen del Rocío, Department of Psychiatry, Universidad de Sevilla, Sevilla, Spain; Instituto de Investigación Sanitaria de Sevilla, IBiS, Spain; Centro Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain
| | - Rosa Ayesa-Arriola
- Marqués de Valdecilla University Hospital, IDIVAL, School of Medicine, University of Cantabria, Santander, Spain; Centro Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain.
| |
Collapse
|
18
|
Li H, Zeng F, Huang C, Pu Q, Thomas ER, Chen Y, Li X. The potential role of glucose metabolism, lipid metabolism, and amino acid metabolism in the treatment of Parkinson's disease. CNS Neurosci Ther 2024; 30:e14411. [PMID: 37577934 PMCID: PMC10848100 DOI: 10.1111/cns.14411] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 08/15/2023] Open
Abstract
PURPOSE OF REVIEW Parkinson's disease (PD) is a common neurodegenerative disease, which can cause progressive deterioration of motor function causing muscle stiffness, tremor, and bradykinesia. In this review, we hope to describe approaches that can improve the life of PD patients through modifications of energy metabolism. RECENT FINDINGS The main pathological features of PD are the progressive loss of nigrostriatal dopaminergic neurons and the production of Lewy bodies. Abnormal aggregation of α-synuclein (α-Syn) leading to the formation of Lewy bodies is closely associated with neuronal dysfunction and degeneration. The main causes of PD are said to be mitochondrial damage, oxidative stress, inflammation, and abnormal protein aggregation. Presence of abnormal energy metabolism is another cause of PD. Many studies have found significant differences between neurodegenerative diseases and metabolic decompensation, which has become a biological hallmark of neurodegenerative diseases. SUMMARY In this review, we highlight the relationship between abnormal energy metabolism (Glucose metabolism, lipid metabolism, and amino acid metabolism) and PD. Improvement of key molecules in glucose metabolism, fat metabolism, and amino acid metabolism (e.g., glucose-6-phosphate dehydrogenase, triglycerides, and levodopa) might be potentially beneficial in PD. Some of these metabolic indicators may serve well during the diagnosis of PD. In addition, modulation of these metabolic pathways may be a potential target for the treatment and prevention of PD.
Collapse
Affiliation(s)
- Hangzhen Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical ScienceSouthwest Medical UniversityLuzhouChina
| | - Fancai Zeng
- Department of Biochemistry and Molecular Biology, School of Basic Medical ScienceSouthwest Medical UniversityLuzhouChina
| | - Cancan Huang
- Department of DermatologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
| | - Qiqi Pu
- Department of Biochemistry and Molecular Biology, School of Basic Medical ScienceSouthwest Medical UniversityLuzhouChina
| | | | - Yan Chen
- Department of DermatologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
| | - Xiang Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical ScienceSouthwest Medical UniversityLuzhouChina
| |
Collapse
|
19
|
de Oliveira J, Moreira ELG, de Bem AF. Beyond cardiovascular risk: Implications of Familial hypercholesterolemia on cognition and brain function. Ageing Res Rev 2024; 93:102149. [PMID: 38056504 DOI: 10.1016/j.arr.2023.102149] [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: 08/08/2023] [Revised: 11/20/2023] [Accepted: 11/30/2023] [Indexed: 12/08/2023]
Abstract
Familial hypercholesterolemia (FH) is a metabolic condition caused mainly by a mutation in the low-density lipoprotein (LDL) receptor gene (LDLR), which is highly prevalent in the population. Besides being an important causative factor of cardiovascular diseases, FH has been considered an early risk factor for Alzheimer's disease. Cognitive and emotional behavioral impairments in LDL receptor knockout (LDLr-/-) mice are associated with neuroinflammation, blood-brain barrier dysfunction, impaired neurogenesis, brain oxidative stress, and mitochondrial dysfunction. Notably, today, LDLr-/- mice, a widely used animal model for studying cardiovascular diseases and atherosclerosis, are also considered an interesting tool for studying dementia. Here, we reviewed the main findings in LDLr-/- mice regarding the relationship between FH and brain dysfunctions and dementia development.
Collapse
Affiliation(s)
- Jade de Oliveira
- Laboratory of investigation on metabolic disorders and neurodegenerative diseases, Department of Biochemistry, Federal University of Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil.
| | - Eduardo Luiz Gasnhar Moreira
- Neuroscience Coworking Lab, Department of Physiological Sciences, Federal University of Santa Catarina, Florianópolis, SC 88040-900, Brazil.
| | - Andreza Fabro de Bem
- Laboratory of Bioenergetics and Metabolism, Department of Physiological Sciences, University of Brasilia, Brasília, Federal District, DF 70910-900, Brazil; Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden; Brazilian National Institute of Science and Technology on Neuroimmunomodulation, Oswaldo Cruz Foundation, Rio de Janeiro, RJ 21040360, Brazil.
| |
Collapse
|
20
|
Tripodi D, Vitarelli F, Spiti S, Leoni V. The Diagnostic Use of the Plasma Quantification of 24S-Hydroxycholesterol and Other Oxysterols in Neurodegenerative Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1440:337-351. [PMID: 38036888 DOI: 10.1007/978-3-031-43883-7_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
Cholesterol regulates fluidity and structure of cellular membranes. The brain is involved in signal transduction, synaptogenesis, and membrane trafficking. An impairment of its metabolism was observed in different neurodegenerative diseases, such as Multiple Sclerosis, Alzheimer, and Huntington diseases. Because of the blood-brain barrier, cholesterol cannot be uptaken from the circulation and all the cholesterol is locally synthetized. The excess cholesterol in neurons is converted into 24S-hydroxycholesterol (24OHC) by the cholesterol 24-hydroxylase (CYP46A1). The plasmatic concentration of 24OHC results in the balance between cerebral production and liver elimination. It is related to the number of metabolically active neurons in the brain. Several factors that affect the brain cholesterol turnover and the liver elimination of oxysterols, the genetic background, nutrition, and lifestyle habits were found to significantly affect plasma levels of 24OHC. Reduced levels of 24OHC were found related to the loss of metabolically active cells and the degree of brain atrophy. The dysfunction of the blood-brain barrier, inflammation, and increased cholesterol turnover might overlap with this progressive reduction giving temporary increased levels of 24OHC.The study of plasma 24OHC is likely to offer an insight into brain cholesterol turnover with a limited diagnostic power.
Collapse
Affiliation(s)
- Domenico Tripodi
- Laboratory of Clinical Pathology, Hospital Pio XI of Desio, ASST-Brianza and Department of Medicine and Surgery, University of Milano Bicocca, Desio, MB, Italy
| | - Federica Vitarelli
- Laboratory of Clinical Pathology, Hospital Pio XI of Desio, ASST-Brianza and Department of Medicine and Surgery, University of Milano Bicocca, Desio, MB, Italy
| | - Simona Spiti
- Laboratory of Clinical Pathology, Hospital Pio XI of Desio, ASST-Brianza and Department of Medicine and Surgery, University of Milano Bicocca, Desio, MB, Italy
| | - Valerio Leoni
- Laboratory of Clinical Pathology, Hospital Pio XI of Desio, ASST-Brianza and Department of Medicine and Surgery, University of Milano Bicocca, Desio, MB, Italy.
| |
Collapse
|
21
|
Zarezade V, Nazeri Z, Azizidoost S, Cheraghzadeh M, Babaahmadi-Rezaei H, Kheirollah A. Paradoxical effect of Aβ on protein levels of ABCA1 in astrocytes, microglia, and neurons isolated from C57BL/6 mice: an in vitro and in silico study to elucidate the effect of Aβ on ABCA1 in the brain cells. J Biomol Struct Dyn 2024; 42:274-287. [PMID: 37105231 DOI: 10.1080/07391102.2023.2201835] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 03/10/2023] [Indexed: 04/29/2023]
Abstract
Impaired cholesterol metabolism has been reported in Alzheimer's disease. Since ABCA1 is one of the main players in the brain's cholesterol homeostasis, here we used the in-vitro and in-silico experiments to investigate the effect of Aβ on ABCA1 protein levels in microglia, astrocytes, and neurons in mice. Microglia, astrocytes, and neurons were cultured and exposed to beta amyloid. ABCA1 in cell lysates was determined by Western blotting, and cholesterol efflux was measured in the conditioned media. Molecular docking, molecular dynamics simulations, and MM-GBSA analysis were conducted to gain a better understanding of the effects of Aβ on ABCA1. In response to Aβ, the protein levels of ABCA1 increase significantly in microglia, astrocytes, and neurons; however, its ability to enhance cholesterol efflux is diminished. Aβ inhibited the function of ABCA1 by obstructing the extracellular tunnel that transports lipids outside the cell, as determined by molecular docking. MD simulation analysis validated these findings. Our results demonstrated that Aβ could increase ABCA1 protein levels in various brain cells, regardless of cell type. Molecular docking, molecular dynamics simulation, and MM-GBSA studies indicate that Aβ has a significant effect on the structural conformation of ABCA1, possibly interfering with its function. We believe that the conformational changes of ABCA1 will inhibit its ability to subsequently release cellular cholesterol. Aβ may obstruct the extracellular tunnel of ABCA1, rendering it less accessible to proteases such as the calpain family, which may explain the increase in ABCA1 levels but decrease in its function.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Vahid Zarezade
- Hyperlipidemia Research Center, Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Zahra Nazeri
- Department of Biochemistry, Medical School, Cellular & Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Shirin Azizidoost
- Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Maryam Cheraghzadeh
- Department of Biochemistry, Medical School, Cellular & Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hossein Babaahmadi-Rezaei
- Hyperlipidemia Research Center, Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Alireza Kheirollah
- Department of Biochemistry, Medical School, Cellular & Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
22
|
Zhou L, Zhang CL, Jiang K, Cheng HY, Xiong WW, Zhu JX. Therapeutic Potential of Danyankang Capsule in High-Fat Diet-Induced Cholelithiasis and Its Impact on Liver FXR Signaling and Gut Microbiota. Biol Pharm Bull 2024; 47:680-691. [PMID: 38522942 DOI: 10.1248/bpb.b24-00052] [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] [Indexed: 03/26/2024]
Abstract
Cholelithiasis, commonly known as gallstones, represents a prevalent hepatobiliary disorder. This study aimed to elucidate the therapeutic role and mechanism of Danyankang capsulein treating cholelithiasis induced by a high-fat diet in C57BL/6 mice. The therapeutical potential of Danyankang was assessed through biochemical analyses, histopathological examinations, protein detection, and 16S rDNA sequencing. A high-fat diet resulted in cholelithiasis manifestation in mice, with discernable abnormal serum biochemical indices and disrupted biliary cholesterol homeostasis. Danyankang treatment notably ameliorated liver inflammation symptoms and rectified serum and liver biochemical abnormalities. Concurrently, it addressed biliary imbalances. Elevated expressions of toll-like receptor 4 (TLR4), nuclear factor-kappaB (NF-κB)/pNF-κB, HMGCR, CYP7A1, and CYP8B1 observed at the inception of cholelithiasis, were notably reduced upon Danyankang administration. Furthermore, 16S rDNA analysis revealed a decline in species number and diversity of the intestinal flora in cholelithiasis-treated mice, while the decline was reversed with Danyankang treatment. Danyankang capsules reduced the abundance of Verrucomicrobiota and increased the abundance of Actinobacteriota and Proteobacteria. In conclusion, the present study demonstrates that Danyankang exerts potent therapeutic efficacy against high-fat diet-induced cholelithiasis. This beneficial outcome is potentially linked to the inhibition of the TLR4/pNF-κB and SHP/CYP7A1/CYP8B1 signaling pathways, as well as the enhancement of intestinal flora species abundance.
Collapse
Affiliation(s)
- Lin Zhou
- Research Center of Traditional Chinese Medicine Resources and Ethnic Medicine, Jiangxi University of Chinese Medicine
| | - Chu-Ling Zhang
- Research Center of Traditional Chinese Medicine Resources and Ethnic Medicine, Jiangxi University of Chinese Medicine
| | - Kun Jiang
- Guizhou Bailing Enterprise Group Pharmaceutical Co., Ltd
| | - Hong-Yu Cheng
- College of Humanities, Jiangxi University of Chinese Medicine
| | - Wen-Wen Xiong
- Medical Clinic, Jiangxi University of Chinese Medicine
| | - Ji-Xiao Zhu
- Research Center of Traditional Chinese Medicine Resources and Ethnic Medicine, Jiangxi University of Chinese Medicine
| |
Collapse
|
23
|
Hu ZL, Yuan YQ, Tong Z, Liao MQ, Yuan SL, Jian Y, Yang JL, Liu WF. Reexamining the Causes and Effects of Cholesterol Deposition in the Brains of Patients with Alzheimer's Disease. Mol Neurobiol 2023; 60:6852-6868. [PMID: 37507575 DOI: 10.1007/s12035-023-03529-y] [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: 10/13/2022] [Accepted: 07/20/2023] [Indexed: 07/30/2023]
Abstract
Alzheimer's disease (AD) is a degenerative disease of the central nervous system. Numerous studies have shown that imbalances in cholesterol homeostasis in the brains of AD patients precede the onset of clinical symptoms. In addition, cholesterol deposition has been observed in the brains of AD patients even though peripheral cholesterol does not enter the brain through the blood‒brain barrier (BBB). Studies have demonstrated that cholesterol metabolism in the brain is associated with many pathological conditions, such as amyloid beta (Aβ) production, Tau protein phosphorylation, oxidative stress, and inflammation. In 2022, some scholars put forward a new hypothesis of AD: the disease involves lipid invasion and its exacerbation of the abnormal metabolism of cholesterol in the brain. In this review, by discussing the latest research progress, the causes and effects of cholesterol retention in the brains of AD patients are analyzed and discussed. Additionally, the possible mechanism through which AD may be improved by targeting cholesterol is described. Finally, we propose that improving the impairments in cholesterol removal observed in the brains of AD patients, instead of further reducing the already impaired cholesterol synthesis in the brain, may be the key to preventing cholesterol deposition and improving the corresponding pathological symptoms.
Collapse
Affiliation(s)
- Ze-Lin Hu
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China
| | - Yang-Qi Yuan
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China
| | - Zhen Tong
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China
| | - Mei-Qing Liao
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China
| | - Shun-Ling Yuan
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China
| | - Ye Jian
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China
| | - Jia-Lun Yang
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China
| | - Wen-Feng Liu
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha, 410012, China.
- Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, Hunan Normal University, Changsha, 410081, China.
| |
Collapse
|
24
|
Gilbert MC, Setayesh T, Wan YJY. The contributions of bacteria metabolites to the development of hepatic encephalopathy. LIVER RESEARCH 2023; 7:296-303. [PMID: 38221945 PMCID: PMC10786625 DOI: 10.1016/j.livres.2022.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Over 20% of mortality during acute liver failure is associated with the development of hepatic encephalopathy (HE). Thus, HE is a complication of acute liver failure with a broad spectrum of neuropsychiatric abnormalities ranging from subclinical alterations to coma. HE is caused by the diversion of portal blood into systemic circulation through portosystemic collateral vessels. Thus, the brain is exposed to intestinal-derived toxic substances. Moreover, the strategies to prevent advancement and improve the prognosis of such a liver-brain disease rely on intestinal microbial modulation. This is supported by the findings that antibiotics such as rifaximin and laxative lactulose can alleviate hepatic cirrhosis and/or prevent HE. Together, the significance of the gut-liver-brain axis in human health warrants attention. This review paper focuses on the roles of bacteria metabolites, mainly ammonia and bile acids (BAs) as well as BA receptors in HE. The literature search conducted for this review included searches for phrases such as BA receptors, BAs, ammonia, farnesoid X receptor (FXR), G protein-coupled bile acid receptor 1 (GPBAR1 or TGR5), sphingosine-1-phosphate receptor 2 (S1PR2), and cirrhosis in conjunction with the phrase hepatic encephalopathy and portosystemic encephalopathy. PubMed, as well as Google Scholar, was the search engines used to find relevant publications.
Collapse
Affiliation(s)
- Miranda Claire Gilbert
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, CA, USA
| | - Tahereh Setayesh
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, CA, USA
| | - Yu-Jui Yvonne Wan
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, CA, USA
| |
Collapse
|
25
|
Constantinescu CC, Brown T, Wang S, Yin W, Barret O, Jennings D, Tauscher J. Clinical Characterization of [ 18F]T-008, a Cholesterol 24-Hydroxylase PET Ligand: Dosimetry, Kinetic Modeling, Variability, and Soticlestat Occupancy. J Nucl Med 2023; 64:1972-1979. [PMID: 37770111 PMCID: PMC10690114 DOI: 10.2967/jnumed.123.265912] [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: 04/20/2023] [Revised: 08/18/2023] [Indexed: 10/03/2023] Open
Abstract
This series of studies characterized [18F]T-008, a PET radiotracer for imaging cholesterol 24-hydroxylase (CH24H), in healthy volunteers (ClinicalTrials.gov identifier NCT02497235). Assessments included radiation dosimetry, kinetic modeling, test-retest variability (TRT) evaluation, and a dose occupancy evaluation using soticlestat, a selective CH24H inhibitor. Soticlestat is currently in phase 3 development for the treatment of seizures in Dravet syndrome and Lennox-Gastaut syndrome. Methods: In the dosimetry study, 5 participants (3 men) underwent serial whole-body scans to estimate organ-absorbed doses and effective doses of [18F]T-008 using OLINDA/EXM 1.1. For the kinetic modeling and TRT study, 6 participants (all men) underwent two 210-min dynamic [18F]T-008 PET scans with arterial blood sampling. The regional total volume of distribution was estimated using a 1-tissue-compartment model, a 2-tissue-compartment model, and Logan graphic analysis. In the dose occupancy study, 11 participants (all men) underwent 120-min scans at baseline and 2 time points (peak and trough) after receiving single oral doses of soticlestat (50-600 mg). The relationship between effect-site soticlestat concentration and brain occupancy was evaluated with a specially developed pharmacokinetic model and a saturable maximal occupancy model. Results: The estimated mean whole-body effective dose was 0.0292 mSv/MBq (SD, 0.00147 mSv/MBq). [18F]T-008 entered the brain rapidly, with a distribution consistent with known CH24H distribution densities. The 2-tissue-compartment model and Logan graphic analysis best described the tracer kinetics. The mean TRT for estimating total volume of distribution was 7%-15%. Single doses of soticlestat in the range 50-600 mg resulted in occupancies of 64%-96% at 2 h and 11%-79% at 24 h. The estimated half-maximal effect-site concentration of soticlestat was 5.52 ng/mL. Conclusion: [18F]T-008 is a suitable PET radiotracer for quantitatively analyzing CH24H in the human brain. Using [18F]T-008 and PET, we demonstrated that soticlestat was brain-penetrant and established target engagement by displacing [18F]T-008 in a dose-dependent manner in the brain.
Collapse
Affiliation(s)
| | - Terry Brown
- Takeda Pharmaceutical Co. Ltd., Cambridge, Massachusetts
| | - Shining Wang
- Takeda Pharmaceutical Co. Ltd., Cambridge, Massachusetts
| | - Wei Yin
- Takeda Pharmaceutical Co. Ltd., Cambridge, Massachusetts
| | | | | | | |
Collapse
|
26
|
Nisar S, Haris M. Neuroimaging genetics approaches to identify new biomarkers for the early diagnosis of autism spectrum disorder. Mol Psychiatry 2023; 28:4995-5008. [PMID: 37069342 PMCID: PMC11041805 DOI: 10.1038/s41380-023-02060-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 03/23/2023] [Accepted: 03/28/2023] [Indexed: 04/19/2023]
Abstract
Autism-spectrum disorders (ASDs) are developmental disabilities that manifest in early childhood and are characterized by qualitative abnormalities in social behaviors, communication skills, and restrictive or repetitive behaviors. To explore the neurobiological mechanisms in ASD, extensive research has been done to identify potential diagnostic biomarkers through a neuroimaging genetics approach. Neuroimaging genetics helps to identify ASD-risk genes that contribute to structural and functional variations in brain circuitry and validate biological changes by elucidating the mechanisms and pathways that confer genetic risk. Integrating artificial intelligence models with neuroimaging data lays the groundwork for accurate diagnosis and facilitates the identification of early diagnostic biomarkers for ASD. This review discusses the significance of neuroimaging genetics approaches to gaining a better understanding of the perturbed neurochemical system and molecular pathways in ASD and how these approaches can detect structural, functional, and metabolic changes and lead to the discovery of novel biomarkers for the early diagnosis of ASD.
Collapse
Affiliation(s)
- Sabah Nisar
- Laboratory of Molecular and Metabolic Imaging, Sidra Medicine, Doha, Qatar
- Department of Diagnostic Imaging, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Mohammad Haris
- Laboratory of Molecular and Metabolic Imaging, Sidra Medicine, Doha, Qatar.
- Center for Advanced Metabolic Imaging in Precision Medicine, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Laboratory Animal Research Center, Qatar University, Doha, Qatar.
| |
Collapse
|
27
|
Gao Y, Ye S, Tang Y, Tong W, Sun S. Brain cholesterol homeostasis and its association with neurodegenerative diseases. Neurochem Int 2023; 171:105635. [PMID: 37949118 DOI: 10.1016/j.neuint.2023.105635] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 10/17/2023] [Accepted: 10/27/2023] [Indexed: 11/12/2023]
Abstract
The brain is the most cholesterol-rich organ in mammals. However, cholesterol metabolism in the brain is completely independent of other tissues due to the presence of the blood-brain barrier (BBB). Neurons, astrocytes and oligodendrocytes are the main cells responsible for cholesterol synthesis in the brain. The cholesterol content in the brain is maintained at a relatively constant level under strict regulation of synthesis, transport, and turnover, that is, brain cholesterol homeostasis. Once this balance is disrupted, neurodegenerative diseases including Alzheimer's disease (AD), Parkinson's disease (PD) and Huntington's disease (HD) ensue. This review summarizes the processes controlling cholesterol homeostasis with respect to the synthesis, transport and turnover of cholesterol in the brain. We further focus on how cholesterol imbalance contributes to neurodegenerative diseases to explore the possibilities to modulate the key steps involved, which will provide clues for the development of therapies for the treatment of central nervous system (CNS) diseases.
Collapse
Affiliation(s)
- Yi Gao
- Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Shiying Ye
- Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yuehong Tang
- Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Wenjuan Tong
- Department of Gynecology and Obstetrics, First Affiliated Hospital, University of South China, Hengyang, Hunan, 421001, China.
| | - Shaowei Sun
- Institute of Pharmacy and Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China.
| |
Collapse
|
28
|
Rani A, Tanwar M, Verma TP, Patra P, Trivedi P, Kumar R, Jha HC. Understanding the role of membrane cholesterol upon Epstein Barr virus infection in astroglial cells. Front Immunol 2023; 14:1192032. [PMID: 37876925 PMCID: PMC10591182 DOI: 10.3389/fimmu.2023.1192032] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 09/21/2023] [Indexed: 10/26/2023] Open
Abstract
Background EBV infection has long been postulated to trigger multiple sclerosis (MS) and anti-EBV antibodies showed a consistent presence in MS patients. Previous reports from our group have shown that the EBV infects different brain cells. Entry of the virus in neuronal cells is assisted by several host factors including membrane cholesterol. By using an inhibitor, methyl-β-cyclodextrin (MβCD), we evaluated the role of membrane cholesterol in EBV infection and pathogenesis. Methodology The membrane cholesterol depleted cells were infected with EBV and its latent genes expression were assessed. Further, EBV-mediated downstream signalling molecules namely STAT3, RIP, NF-kB and TNF-α levels was checked at protein level along with spatial (periphery and nucleus) and temporal changes in biomolecular fingerprints with Raman microspectroscopy (RS). Results Upon treatment with MβCD, lmp1 and lmp2a suggested significant downregulation compared to EBV infection. Downstream molecules like STAT3 and RIP, exhibited a decrease in protein levels temporally upon exposure to MβCD while NF-kB levels were found to be increased. Further, the intensity of the Raman spectra exhibited an increase in triglycerides and fatty acids in the cytoplasm of EBV-infected LN-229 cells compared to MβCD+EBV. Likewise, the Raman peak width of cholesterol, lipid and fatty acids were found to be reduced in EBV-infected samples indicates elevation in the cholesterol specific moieties. In contrast, an opposite pattern was observed in the nucleus. Moreover, the ingenuity pathway analysis revealed protein molecules such as VLDLR, MBP and APP that are associated with altered profile of cholesterol, fatty acids and triglycerides with infection-related CNS disorders. Conclusion Taken together, our results underline the important role of membrane cholesterol over EBV entry/pathogenesis in astroglia cells which further trigger/exacerbate virus-associated neuropathologies. These results likely to aid into the prognosis of neurological disease like MS.
Collapse
Affiliation(s)
- Annu Rani
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology, Indore, India
| | - Manushree Tanwar
- Materials and Device Laboratory, Department of Physics, Indian Institute of Technology, Indore, India
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA, United States
| | - Tarun Prakash Verma
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology, Indore, India
| | - Priyanka Patra
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology, Indore, India
| | - Pankaj Trivedi
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Rajesh Kumar
- Materials and Device Laboratory, Department of Physics, Indian Institute of Technology, Indore, India
| | - Hem Chandra Jha
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology, Indore, India
| |
Collapse
|
29
|
Alrosan AZ, Heilat GB, Al Subeh ZY, Alrosan K, Alrousan AF, Abu-Safieh AK, Alabdallat NS. The effects of statin therapy on brain tumors, particularly glioma: a review. Anticancer Drugs 2023; 34:985-994. [PMID: 37466094 PMCID: PMC10501357 DOI: 10.1097/cad.0000000000001533] [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: 06/21/2023] [Revised: 06/21/2023] [Indexed: 07/20/2023]
Abstract
Brain tumors account for less than 2% of all malignancies. However, they are associated with the highest morbidity and mortality rates among all solid tumors. The most common malignant primary brain tumors are glioma or glioblastoma (GBM), which have a median survival time of about 14 months, often suffer from recurrence after a few months following treatment, and pose a therapeutic challenge. Despite recent therapeutic advances, the prognosis for glioma patients is poor when treated with modern therapies, including chemotherapy, surgery, radiation, or a combination of these. Therefore, discovering a new target to treat brain tumors, particularly glioma, might be advantageous in raising progression-free survival and overall survival (OS) rates. Statins, also known as competitive HMG-CoA reductase inhibitors, are effective medications for reducing cholesterol and cardiovascular risk. The use of statins prior to and during other cancer treatments appears to enhance patient outcomes according to preclinical studies. After surgical resection followed by concurrent radiation and treatment, OS for patients with GBM is only about a year. Statins have recently emerged as potential adjuvant medications for treating GBM due to their ability to inhibit cell growth, survival, migration, metastasis, inflammation, angiogenesis, and increase apoptosis in-vitro and in-vivo studies. Whether statins enhance clinical outcomes, such as patient survival in GBM, is still debatable. This study aimed to explore the effects of statin therapy in the context of cancer treatment, with a particular focus on GBM.
Collapse
Affiliation(s)
- Amjad Z. Alrosan
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmaceutical Sciences, The Hashemite University, Zarqa
| | - Ghaith B. Heilat
- Department of General Surgery and Urology, Faculty of Medicine, The Jordan University of Science and Technology
| | - Zeinab Y. Al Subeh
- Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmacy, The Jordan University of Science and Technology
| | - Khaled Alrosan
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmaceutical Sciences, The Hashemite University, Zarqa
| | - Alaa F. Alrousan
- Doctor of Pharmacy, Faculty of Pharmacy, The Jordan University of Science and Technology, Irbid
| | - Amro K. Abu-Safieh
- Faculty of Pharmaceutical Sciences, The Hashemite University, Zarqa, Jordan
| | | |
Collapse
|
30
|
Freiría-Martínez L, Iglesias-Martínez-Almeida M, Rodríguez-Jamardo C, Rivera-Baltanás T, Comís-Tuche M, Rodrígues-Amorím D, Fernández-Palleiro P, Blanco-Formoso M, Álvarez-Chaver P, Diz-Chaves Y, Gonzalez-Freiria N, Martín-Forero-Maestre M, Fernández-Feijoo CD, Suárez-Albo M, Fernández-Lorenzo JR, Guisán AC, Olivares JM, Spuch C. Proteomic analysis of exosomes derived from human mature milk and colostrum of mothers with term, late preterm, or very preterm delivery. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2023; 15:4905-4917. [PMID: 37718950 DOI: 10.1039/d3ay01114c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2023]
Abstract
The growth and development of the human brain is a long and complex process that requires a precise sequence of genetic and molecular events. This begins in the third week of gestation with the differentiation of neural progenitor cells and extends at least until late adolescence, possibly for life. One of the defects of this development is that we know very little about the signals that modulate this sequence of events. The first 3 years of life, during breastfeeding, is one of the critical periods in brain development. In these first years of life, it is believed that neurodevelopmental problems may be the molecular causes of mental disorders. Therefore, we herein propose a new hypothesis, according to which the chemical signals that could modulate this entire complex sequence of events appear in this early period, and the molecular level study of human breast milk and colostrum of mothers who give birth to children in different gestation periods could give us information on proteins influencing this process. In this work, we collected milk and colostrum samples (term, late preterm and moderate/very preterm) and exosomes were isolated. The samples of exosomes and complete milk from each fraction were analyzed by LC-ESI-MS/MS. In this work, we describe proteins in the different fractions of mature milk and colostrum of mothers with term, late preterm, or very preterm delivery, which could be involved in the regulation of the nervous system by their functions. We describe how they differ in different types of milk, paving the way for the investigation of possible new neuroregulatory pathways as possible candidates to modulate the nervous system.
Collapse
Affiliation(s)
- Luis Freiría-Martínez
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, 36312, Spain.
- University of Vigo, Vigo, 36310, Spain
| | - Marta Iglesias-Martínez-Almeida
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, 36312, Spain.
- University of Vigo, Vigo, 36310, Spain
| | - Cynthia Rodríguez-Jamardo
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, 36312, Spain.
- University of Vigo, Vigo, 36310, Spain
| | - Tania Rivera-Baltanás
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, 36312, Spain.
- CIBERSAM, Madrid, 28029, Spain.
| | - María Comís-Tuche
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, 36312, Spain.
| | - Daniela Rodrígues-Amorím
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, 36312, Spain.
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Patricia Fernández-Palleiro
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, 36312, Spain.
| | - María Blanco-Formoso
- Department of Physical Chemistry, Singular Center for Biomedical Research (CINBIO), Universidade de Vigo, Vigo, 36310, Spain
| | - Paula Álvarez-Chaver
- Structural Determination, Proteomic and Genomic Service, CACTI, University of Vigo, Vigo, Spain
| | - Yolanda Diz-Chaves
- Laboratory of Endocrinology, Singular Center for Biomedical Research (CINBIO), Universidade de Vigo, 36310 Vigo, Spain
| | | | | | | | - María Suárez-Albo
- Neonatal Intensive Care Unit, Alvaro Cunqueiro Hospital, Vigo, 36312, Spain
| | | | | | - Jose Manuel Olivares
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, 36312, Spain.
- CIBERSAM, Madrid, 28029, Spain.
| | - Carlos Spuch
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, 36312, Spain.
- CIBERSAM, Madrid, 28029, Spain.
| |
Collapse
|
31
|
Xie W, Li J, Du H, Xia J. Causal relationship between PCSK9 inhibitor and autoimmune diseases: a drug target Mendelian randomization study. Arthritis Res Ther 2023; 25:148. [PMID: 37580807 PMCID: PMC10424393 DOI: 10.1186/s13075-023-03122-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 07/19/2023] [Indexed: 08/16/2023] Open
Abstract
BACKGROUND In addition to decreasing the level of cholesterol, proprotein convertase subtilis kexin 9 (PCSK9) inhibitor has pleiotropic effects, including immune regulation. However, the impact of PCSK9 on autoimmune diseases is controversial. Therefore, we used drug target Mendelian randomization (MR) analysis to investigate the effect of PCSK9 inhibitor on different autoimmune diseases. METHODS We collected single nucleotide polymorphisms (SNPs) of PCSK9 from published genome-wide association studies statistics and conducted drug target MR analysis to detect the causal relationship between PCSK9 inhibitor and the risk of autoimmune diseases. 3-Hydroxy-3-methylglutaryl-assisted enzyme A reductase (HMGCR) inhibitor, the drug target of statin, was used to compare the effect with that of PCSK9 inhibitor. With the risk of coronary heart disease as a positive control, primary outcomes included the risk of systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), myasthenia gravis (MG), multiple sclerosis (MS), asthma, Crohn's disease (CD), ulcerative colitis (UC), and type 1 diabetes (T1D). RESULTS PCSK9 inhibitor significantly reduced the risk of SLE (OR [95%CI] = 0.47 [0.30 to 0.76], p = 1.74 × 10-3) but increased the risk of asthma (OR [95%CI] = 1.15 [1.03 to 1.29], p = 1.68 × 10-2) and CD (OR [95%CI] = 1.38 [1.05 to 1.83], p = 2.28 × 10-2). In contrast, HMGCR inhibitor increased the risk of RA (OR [95%CI] = 1.58 [1.19 to 2.11], p = 1.67 × 10-3), asthma (OR [95%CI] = 1.21 [1.04 to 1.40], p = 1.17 × 10-2), and CD (OR [95%CI] = 1.60 [1.08 to 2.39], p = 2.04 × 10-2). CONCLUSIONS PCSK9 inhibitor significantly reduced the risk of SLE but increased the risk of asthma and CD. In contrast, HMGCR inhibitor may be a risk factor for RA, asthma, and CD.
Collapse
Affiliation(s)
- Weijia Xie
- Department of Neurology, Xiangya Hospital, Central South University, 87# Xiangya Road, Changsha, Hunan, China
| | - Jiaxin Li
- Department of Neurology, Xiangya Hospital, Central South University, 87# Xiangya Road, Changsha, Hunan, China
| | - Hao Du
- Department of Neurology, Xiangya Hospital, Central South University, 87# Xiangya Road, Changsha, Hunan, China
| | - Jian Xia
- Department of Neurology, Xiangya Hospital, Central South University, 87# Xiangya Road, Changsha, Hunan, China.
- Clinical Research Center for Cerebrovascular Disease of Hunan Province, Central South University, Changsha, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
32
|
Deng R, Medico-Salsench E, Nikoncuk A, Ramakrishnan R, Lanko K, Kühn NA, van der Linde HC, Lor-Zade S, Albuainain F, Shi Y, Yousefi S, Capo I, van den Herik EM, van Slegtenhorst M, van Minkelen R, Geeven G, Mulder MT, Ruijter GJG, Lütjohann D, Jacobs EH, Houlden H, Pagnamenta AT, Metcalfe K, Jackson A, Banka S, De Simone L, Schwaede A, Kuntz N, Palculict TB, Abbas S, Umair M, AlMuhaizea M, Colak D, AlQudairy H, Alsagob M, Pereira C, Trunzo R, Karageorgou V, Bertoli-Avella AM, Bauer P, Bouman A, Hoefsloot LH, van Ham TJ, Issa M, Zaki MS, Gleeson JG, Willemsen R, Kaya N, Arold ST, Maroofian R, Sanderson LE, Barakat TS. AMFR dysfunction causes autosomal recessive spastic paraplegia in human that is amenable to statin treatment in a preclinical model. Acta Neuropathol 2023; 146:353-368. [PMID: 37119330 PMCID: PMC10328903 DOI: 10.1007/s00401-023-02579-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/17/2023] [Accepted: 04/18/2023] [Indexed: 05/01/2023]
Abstract
Hereditary spastic paraplegias (HSP) are rare, inherited neurodegenerative or neurodevelopmental disorders that mainly present with lower limb spasticity and muscle weakness due to motor neuron dysfunction. Whole genome sequencing identified bi-allelic truncating variants in AMFR, encoding a RING-H2 finger E3 ubiquitin ligase anchored at the membrane of the endoplasmic reticulum (ER), in two previously genetically unexplained HSP-affected siblings. Subsequently, international collaboration recognized additional HSP-affected individuals with similar bi-allelic truncating AMFR variants, resulting in a cohort of 20 individuals from 8 unrelated, consanguineous families. Variants segregated with a phenotype of mainly pure but also complex HSP consisting of global developmental delay, mild intellectual disability, motor dysfunction, and progressive spasticity. Patient-derived fibroblasts, neural stem cells (NSCs), and in vivo zebrafish modeling were used to investigate pathomechanisms, including initial preclinical therapy assessment. The absence of AMFR disturbs lipid homeostasis, causing lipid droplet accumulation in NSCs and patient-derived fibroblasts which is rescued upon AMFR re-expression. Electron microscopy indicates ER morphology alterations in the absence of AMFR. Similar findings are seen in amfra-/- zebrafish larvae, in addition to altered touch-evoked escape response and defects in motor neuron branching, phenocopying the HSP observed in patients. Interestingly, administration of FDA-approved statins improves touch-evoked escape response and motor neuron branching defects in amfra-/- zebrafish larvae, suggesting potential therapeutic implications. Our genetic and functional studies identify bi-allelic truncating variants in AMFR as a cause of a novel autosomal recessive HSP by altering lipid metabolism, which may potentially be therapeutically modulated using precision medicine with statins.
Collapse
Affiliation(s)
- Ruizhi Deng
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Whole Genome Sequencing Implementation and Research Task Force, Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Eva Medico-Salsench
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Anita Nikoncuk
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Reshmi Ramakrishnan
- Bioscience Program, Biological and Environmental Science and Engineering Division, Computational Bioscience Research Center, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900 Saudi Arabia
| | - Kristina Lanko
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Nikolas A. Kühn
- Department of Cell Biology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Herma C. van der Linde
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Sarah Lor-Zade
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Fatimah Albuainain
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Yuwei Shi
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Soheil Yousefi
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Whole Genome Sequencing Implementation and Research Task Force, Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Ivan Capo
- Department for Histology and Embryology, Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia
| | | | - Marjon van Slegtenhorst
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Rick van Minkelen
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Geert Geeven
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Whole Genome Sequencing Implementation and Research Task Force, Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Monique T. Mulder
- Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - George J. G. Ruijter
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Dieter Lütjohann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Edwin H. Jacobs
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Henry Houlden
- Department of Neuromuscular Disorders, UCL Queen Square Institute of Neurology, London, UK
| | - Alistair T. Pagnamenta
- NIHR Biomedical Research Centre, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Kay Metcalfe
- Manchester Centre for Genomic Medicine, St Mary’s Hospital, Health Innovation Manchester, Manchester University Foundation NHS Trust, Manchester, UK
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PL UK
| | - Adam Jackson
- Manchester Centre for Genomic Medicine, St Mary’s Hospital, Health Innovation Manchester, Manchester University Foundation NHS Trust, Manchester, UK
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PL UK
| | - Siddharth Banka
- Manchester Centre for Genomic Medicine, St Mary’s Hospital, Health Innovation Manchester, Manchester University Foundation NHS Trust, Manchester, UK
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PL UK
| | - Lenika De Simone
- Division of Neurology, Division of Genetics, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, USA
| | - Abigail Schwaede
- Division of Neurology, Division of Genetics, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, USA
| | - Nancy Kuntz
- Division of Neurology, Division of Genetics, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, USA
| | | | - Safdar Abbas
- Department of Biological Science, Dartmouth College, Hanover, NH USA
| | - Muhammad Umair
- Medical Genomics Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
- Department of Life Sciences, School of Science, University of Management and Technology (UMT), Lahore, Pakistan
| | - Mohammed AlMuhaizea
- Neuroscience Centre, King Faisal Specialist Hospital and Research Centre (KFSHRC), MBC: 76, Riyadh, 11211 Saudi Arabia
| | - Dilek Colak
- Molecular Oncology Department, King Faisal Specialist Hospital and Research Centre (KFSHRC), MBC: 03, Riyadh, 11211 Saudi Arabia
| | - Hanan AlQudairy
- Translational Genomics Department, Center for Genomics Medicine, King Faisal Specialist Hospital and Research Centre, MBC: 26, PO Box: 3354, Riyadh, 11211 Saudi Arabia
| | - Maysoon Alsagob
- Translational Genomics Department, Center for Genomics Medicine, King Faisal Specialist Hospital and Research Centre, MBC: 26, PO Box: 3354, Riyadh, 11211 Saudi Arabia
- Applied Genomics Technologies Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh, Saudi Arabia
| | | | | | | | | | | | - Arjan Bouman
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Lies H. Hoefsloot
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Whole Genome Sequencing Implementation and Research Task Force, Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Tjakko J. van Ham
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Whole Genome Sequencing Implementation and Research Task Force, Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Mahmoud Issa
- Clinical Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, Egypt
| | - Maha S. Zaki
- Clinical Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, Egypt
| | - Joseph G. Gleeson
- Departments of Neurosciences and Pediatrics, Howard Hughes Medical Institute, University of California, Rady Children’s Institute for Genomic Medicine, San Diego, USA
| | - Rob Willemsen
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Namik Kaya
- Translational Genomics Department, Center for Genomics Medicine, King Faisal Specialist Hospital and Research Centre, MBC: 26, PO Box: 3354, Riyadh, 11211 Saudi Arabia
| | - Stefan T. Arold
- Bioscience Program, Biological and Environmental Science and Engineering Division, Computational Bioscience Research Center, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900 Saudi Arabia
- Centre de Biologie Structurale, CNRS, INSERM, Université de Montpellier, 34090 Montpellier, France
| | - Reza Maroofian
- Department of Neuromuscular Disorders, UCL Queen Square Institute of Neurology, London, UK
| | - Leslie E. Sanderson
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Tahsin Stefan Barakat
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Whole Genome Sequencing Implementation and Research Task Force, Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Discovery Unit, Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
33
|
Gunta SP, O'Keefe JH, O'Keefe EL, Lavie CJ. PCSK9 inhibitor, ezetimibe, and bempedoic acid: Evidence-based therapies for statin-intolerant patients. Prog Cardiovasc Dis 2023; 79:12-18. [PMID: 36871887 DOI: 10.1016/j.pcad.2023.02.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 02/21/2023] [Indexed: 03/07/2023]
Abstract
Statins are first-line therapy for treating dyslipidemia because of their low-density lipoprotein cholesterol (LDL-C) lowering efficacy, superior event-reduction data and unrivaled cost-effectiveness. Yet, many people are intolerant of statins, whether due to true adverse events or the nocebo effect, so within one year about two-thirds of primary prevention patients and one-third of secondary prevention patients are no longer taking their prescription. Statins still dominate this landscape, but other agents, often used in combination, potently reduce LDL-C levels, regress atherosclerosis and lower risk of major adverse cardiovascular events (MACE). Ezetimibe lowers LDL-C by reducing intestinal absorption of cholesterol. Proprotein convertase subtilisin/kexin type 9 inhibitors (PCSK9i) lower LDL-C by increasing the number and durability of hepatic LDL receptors. Bempedoic acid reduces hepatic cholesterol synthesis. Ezetimibe, PCSK9i and bempedoic are evidence-based, non-statin therapies that synergistically lower LDL-C and reduce risk of MACE; they also have benign side-effect profiles and are generally well tolerated.
Collapse
Affiliation(s)
- Satya Preetham Gunta
- Saint Luke's Mid America Heart Institute and University of Missouri-Kansas City, Kansas City, MO, United States of America
| | - James H O'Keefe
- Saint Luke's Mid America Heart Institute and University of Missouri-Kansas City, Kansas City, MO, United States of America.
| | - Evan L O'Keefe
- Tulane Medical Center, New Orleans, LA, United States of America
| | - Carl J Lavie
- John Ochsner Heart and Vascular Institute, Ochsner Clinical School, University of Queensland School of Medicine, New Orleans, LA, United States of America
| |
Collapse
|
34
|
Barmpagiannos K, Theotokis P, Petratos S, Pagnin M, Einstein O, Kesidou E, Boziki M, Artemiadis A, Bakirtzis C, Grigoriadis N. The Diversity of Astrocyte Activation during Multiple Sclerosis: Potential Cellular Targets for Novel Disease Modifying Therapeutics. Healthcare (Basel) 2023; 11:healthcare11111585. [PMID: 37297725 DOI: 10.3390/healthcare11111585] [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: 04/19/2023] [Revised: 05/19/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023] Open
Abstract
Neuroglial cells, and especially astrocytes, constitute the most varied group of central nervous system (CNS) cells, displaying substantial diversity and plasticity during development and in disease states. The morphological changes exhibited by astrocytes during the acute and chronic stages following CNS injury can be characterized more precisely as a dynamic continuum of astrocytic reactivity. Different subpopulations of reactive astrocytes may be ascribed to stages of degenerative progression through their direct pathogenic influence upon neurons, neuroglia, the blood-brain barrier, and infiltrating immune cells. Multiple sclerosis (MS) constitutes an autoimmune demyelinating disease of the CNS. Despite the previously held notion that reactive astrocytes purely form the structured glial scar in MS plaques, their continued multifaceted participation in neuroinflammatory outcomes and oligodendrocyte and neuronal function during chronicity, suggest that they may be an integral cell type that can govern the pathophysiology of MS. From a therapeutic-oriented perspective, astrocytes could serve as key players to limit MS progression, once the integral astrocyte-MS relationship is accurately identified. This review aims toward delineating the current knowledge, which is mainly focused on immunomodulatory therapies of the relapsing-remitting form, while shedding light on uncharted approaches of astrocyte-specific therapies that could constitute novel, innovative applications once the role of specific subgroups in disease pathogenesis is clarified.
Collapse
Affiliation(s)
- Konstantinos Barmpagiannos
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, Aristotle University of Thessaloniki, 54621 Thessaloniki, Greece
| | - Paschalis Theotokis
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, Aristotle University of Thessaloniki, 54621 Thessaloniki, Greece
| | - Steven Petratos
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Maurice Pagnin
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC 3004, Australia
| | - Ofira Einstein
- Department of Physical Therapy, Faculty of Health Sciences, Ariel University, Ariel 40700, Israel
| | - Evangelia Kesidou
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, Aristotle University of Thessaloniki, 54621 Thessaloniki, Greece
| | - Marina Boziki
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, Aristotle University of Thessaloniki, 54621 Thessaloniki, Greece
| | | | - Christos Bakirtzis
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, Aristotle University of Thessaloniki, 54621 Thessaloniki, Greece
| | - Nikolaos Grigoriadis
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, Aristotle University of Thessaloniki, 54621 Thessaloniki, Greece
| |
Collapse
|
35
|
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: 1.5] [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
|
36
|
Bellomo G, Paciotti S, Concha-Marambio L, Rizzo D, Wojdaƚa AL, Chiasserini D, Gatticchi L, Cerofolini L, Giuntini S, De Luca CMG, Ma Y, Farris CM, Pieraccini G, Bologna S, Filidei M, Ravera E, Lelli M, Moda F, Fragai M, Parnetti L, Luchinat C. Cerebrospinal fluid lipoproteins inhibit α-synuclein aggregation by interacting with oligomeric species in seed amplification assays. Mol Neurodegener 2023; 18:20. [PMID: 37005644 PMCID: PMC10068178 DOI: 10.1186/s13024-023-00613-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 03/12/2023] [Indexed: 04/04/2023] Open
Abstract
BACKGROUND Aggregation of α-synuclein (α-syn) is a prominent feature of Parkinson's disease (PD) and other synucleinopathies. Currently, α-syn seed amplification assays (SAAs) using cerebrospinal fluid (CSF) represent the most promising diagnostic tools for synucleinopathies. However, CSF itself contains several compounds that can modulate the aggregation of α-syn in a patient-dependent manner, potentially undermining unoptimized α-syn SAAs and preventing seed quantification. METHODS In this study, we characterized the inhibitory effect of CSF milieu on detection of α-syn aggregates by means of CSF fractionation, mass spectrometry, immunoassays, transmission electron microscopy, solution nuclear magnetic resonance spectroscopy, a highly accurate and standardized diagnostic SAA, and different in vitro aggregation conditions to evaluate spontaneous aggregation of α-syn. RESULTS We found the high-molecular weight fraction of CSF (> 100,000 Da) to be highly inhibitory on α-syn aggregation and identified lipoproteins to be the main drivers of this effect. Direct interaction between lipoproteins and monomeric α-syn was not detected by solution nuclear magnetic resonance spectroscopy, on the other hand we observed lipoprotein-α-syn complexes by transmission electron microscopy. These observations are compatible with hypothesizing an interaction between lipoproteins and oligomeric/proto-fibrillary α-syn intermediates. We observed significantly slower amplification of α-syn seeds in PD CSF when lipoproteins were added to the reaction mix of diagnostic SAA. Additionally, we observed a decreased inhibition capacity of CSF on α-syn aggregation after immunodepleting ApoA1 and ApoE. Finally, we observed that CSF ApoA1 and ApoE levels significantly correlated with SAA kinetic parameters in n = 31 SAA-negative control CSF samples spiked with preformed α-syn aggregates. CONCLUSIONS Our results describe a novel interaction between lipoproteins and α-syn aggregates that inhibits the formation of α-syn fibrils and could have relevant implications. Indeed, the donor-specific inhibition of CSF on α-syn aggregation explains the lack of quantitative results from analysis of SAA-derived kinetic parameters to date. Furthermore, our data show that lipoproteins are the main inhibitory components of CSF, suggesting that lipoprotein concentration measurements could be incorporated into data analysis models to eliminate the confounding effects of CSF milieu on α-syn quantification efforts.
Collapse
Affiliation(s)
- Giovanni Bellomo
- Laboratory of Clinical Neurochemistry, Section of Neurology, Department of Medicine and Surgery, University of Perugia, Piazzale Lucio Severi 1/8, 06132, Perugia, Italy.
| | - Silvia Paciotti
- Laboratory of Clinical Neurochemistry, Section of Neurology, Department of Medicine and Surgery, University of Perugia, Piazzale Lucio Severi 1/8, 06132, Perugia, Italy
| | - Luis Concha-Marambio
- R&D Unit, Amprion Inc, 11095 Flintkote Av., San Diego, San Diego, CA, 92121, USA
| | - Domenico Rizzo
- Magnetic Resonance Center (CERM), University of Florence, Via Luigi Sacconi 6, 50019, Sesto Fiorentino, Italy
- Department of Chemistry "Ugo Schiff", University of Florence, Via Della Lastruccia 3, 50019, Sesto Fiorentino, Italy
| | - Anna Lidia Wojdaƚa
- Laboratory of Clinical Neurochemistry, Section of Neurology, Department of Medicine and Surgery, University of Perugia, Piazzale Lucio Severi 1/8, 06132, Perugia, Italy
| | - Davide Chiasserini
- Section of Physiology and Biochemistry, Department of Medicine and Surgery, University of Perugia, Piazzale Lucio Severi 1/8, 06132, PerugiaPerugia, Italy
| | - Leonardo Gatticchi
- Section of Physiology and Biochemistry, Department of Medicine and Surgery, University of Perugia, Piazzale Lucio Severi 1/8, 06132, PerugiaPerugia, Italy
| | - Linda Cerofolini
- Magnetic Resonance Center (CERM), University of Florence, Via Luigi Sacconi 6, 50019, Sesto Fiorentino, Italy
- Consorzio Interuniversitario Risonanze Magnetiche Metallo Proteine (CIRMMP), Via Luigi Sacconi 6, 50019, Sesto Fiorentino, Italy
| | - Stefano Giuntini
- Magnetic Resonance Center (CERM), University of Florence, Via Luigi Sacconi 6, 50019, Sesto Fiorentino, Italy
| | - Chiara Maria Giulia De Luca
- Division of Neurology 5 and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, 20133, Milan, Italy
| | - Yihua Ma
- R&D Unit, Amprion Inc, 11095 Flintkote Av., San Diego, San Diego, CA, 92121, USA
| | - Carly M Farris
- R&D Unit, Amprion Inc, 11095 Flintkote Av., San Diego, San Diego, CA, 92121, USA
| | - Giuseppe Pieraccini
- Department of Health Sciences, CISM Mass Spectrometry Centre, University of Florence, Viale Gaetano Pieraccini 6, 50139, Florence, Italy
| | - Sara Bologna
- Magnetic Resonance Center (CERM), University of Florence, Via Luigi Sacconi 6, 50019, Sesto Fiorentino, Italy
| | - Marta Filidei
- Laboratory of Clinical Neurochemistry, Section of Neurology, Department of Medicine and Surgery, University of Perugia, Piazzale Lucio Severi 1/8, 06132, Perugia, Italy
| | - Enrico Ravera
- Magnetic Resonance Center (CERM), University of Florence, Via Luigi Sacconi 6, 50019, Sesto Fiorentino, Italy
- Department of Chemistry "Ugo Schiff", University of Florence, Via Della Lastruccia 3, 50019, Sesto Fiorentino, Italy
- Consorzio Interuniversitario Risonanze Magnetiche Metallo Proteine (CIRMMP), Via Luigi Sacconi 6, 50019, Sesto Fiorentino, Italy
| | - Moreno Lelli
- Magnetic Resonance Center (CERM), University of Florence, Via Luigi Sacconi 6, 50019, Sesto Fiorentino, Italy
- Department of Chemistry "Ugo Schiff", University of Florence, Via Della Lastruccia 3, 50019, Sesto Fiorentino, Italy
- Consorzio Interuniversitario Risonanze Magnetiche Metallo Proteine (CIRMMP), Via Luigi Sacconi 6, 50019, Sesto Fiorentino, Italy
| | - Fabio Moda
- Division of Neurology 5 and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, 20133, Milan, Italy
| | - Marco Fragai
- Magnetic Resonance Center (CERM), University of Florence, Via Luigi Sacconi 6, 50019, Sesto Fiorentino, Italy
- Department of Chemistry "Ugo Schiff", University of Florence, Via Della Lastruccia 3, 50019, Sesto Fiorentino, Italy
- Consorzio Interuniversitario Risonanze Magnetiche Metallo Proteine (CIRMMP), Via Luigi Sacconi 6, 50019, Sesto Fiorentino, Italy
| | - Lucilla Parnetti
- Laboratory of Clinical Neurochemistry, Section of Neurology, Department of Medicine and Surgery, University of Perugia, Piazzale Lucio Severi 1/8, 06132, Perugia, Italy
| | - Claudio Luchinat
- Magnetic Resonance Center (CERM), University of Florence, Via Luigi Sacconi 6, 50019, Sesto Fiorentino, Italy.
- Department of Chemistry "Ugo Schiff", University of Florence, Via Della Lastruccia 3, 50019, Sesto Fiorentino, Italy.
- Consorzio Interuniversitario Risonanze Magnetiche Metallo Proteine (CIRMMP), Via Luigi Sacconi 6, 50019, Sesto Fiorentino, Italy.
| |
Collapse
|
37
|
Galkina OV, Vetrovoy OV, Krasovskaya IE, Eschenko ND. Role of Lipids in Regulation of Neuroglial Interactions. BIOCHEMISTRY. BIOKHIMIIA 2023; 88:337-352. [PMID: 37076281 DOI: 10.1134/s0006297923030045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 01/24/2023] [Accepted: 01/26/2023] [Indexed: 03/28/2023]
Abstract
Lipids comprise an extremely heterogeneous group of compounds that perform a wide variety of biological functions. Traditional view of lipids as important structural components of the cell and compounds playing a trophic role is currently being supplemented by information on the possible participation of lipids in signaling, not only intracellular, but also intercellular. The review article discusses current data on the role of lipids and their metabolites formed in glial cells (astrocytes, oligodendrocytes, microglia) in communication of these cells with neurons. In addition to metabolic transformations of lipids in each type of glial cells, special attention is paid to the lipid signal molecules (phosphatidic acid, arachidonic acid and its metabolites, cholesterol, etc.) and the possibility of their participation in realization of synaptic plasticity, as well as in other possible mechanisms associated with neuroplasticity. All these new data can significantly expand our knowledge about the regulatory functions of lipids in neuroglial relationships.
Collapse
Affiliation(s)
- Olga V Galkina
- Biochemistry Department, Faculty of Biology, Saint-Petersburg State University, St. Petersburg, 199034, Russia.
| | - Oleg V Vetrovoy
- Biochemistry Department, Faculty of Biology, Saint-Petersburg State University, St. Petersburg, 199034, Russia
- Pavlov Institute of Physiology, Russian Academy of Sciences, St. Petersburg, 199034, Russia
| | - Irina E Krasovskaya
- Biochemistry Department, Faculty of Biology, Saint-Petersburg State University, St. Petersburg, 199034, Russia
| | - Nataliya D Eschenko
- Biochemistry Department, Faculty of Biology, Saint-Petersburg State University, St. Petersburg, 199034, Russia
| |
Collapse
|
38
|
Escudero-Martínez I, Thorén M, Matusevicius M, Cooray C, Zini A, Roffe C, Toni D, Tsivgoulis G, Ringleb P, Wahlgren N, Ahmed N. Association of cholesterol levels with hemorrhagic transformation and cerebral edema after reperfusion therapies. Eur Stroke J 2022; 8:294-300. [PMID: 37021184 PMCID: PMC10069196 DOI: 10.1177/23969873221148229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 12/13/2022] [Indexed: 12/30/2022] Open
Abstract
Background: The association between cholesterol levels and cerebral edema (CED) or hemorrhagic transformation (HT) as an expressions of blood-brain barrier (BBB) dysfunction after ischemic stroke is not well established. The aim of this study is to determine the association of total cholesterol (TC) levels with the incidence of HT and CED after reperfusion therapies. Methods: We analyzed SITS Thrombolysis and Thrombectomy Registry data from January 2011 to December 2017. We identified patients with data on TC levels at baseline. TC values were categorized in three groups (reference group ⩾200 mg/dl). The two primary outcomes were any parenchymal hemorrhage (PH) and moderate to severe CED on follow up imaging. Secondary outcomes included death and functional independence (mRS 0–2) at 3 months. Multivariable logistic regression analysis adjusted for baseline factors including statin pretreatment was used to assess the association between TC levels and outcomes. Results: Of 35,314 patients with available information on TC levels at baseline, 3372 (9.5%) presented with TC levels ⩽130 mg/dl, 8203 (23.2%) with TC 130–200 mg/dl and 23,739 (67.3%) with TC ⩾ 200 mg/dl. In the adjusted analyses, TC level as continuous variable was inversely associated with moderate to severe CED (OR 0.99, 95% CI 0.99–1.00, p = 0.025) and as categorical variable lower TC levels were associated with a higher risk of moderate to severe CED (aOR 1.24, 95% CI 1.10–1.40, p = 0.003). TC levels were not associated with any PH, functional independence, and mortality at 3 months. Conclusions: Our findings indicate an independent association between low levels of TC and higher odds of moderate/severe CED. Further studies are needed to confirm these findings.
Collapse
Affiliation(s)
- Irene Escudero-Martínez
- Department of Neurology, Hospital Universitari i Poltècnic La Fe, Valencia, Spain
- Neurovascular Research Laboratory, Instituto de Biomedicina de Sevilla-IBiS, Sevilla, Spain
| | - Magnus Thorén
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neurology, Danderyd Hospital, Stockholm, Sweden
| | - Marius Matusevicius
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Charith Cooray
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Andrea Zini
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Department of Neurology and Stroke Center, Maggiore Hospital, Bologna, Italy
| | - Christine Roffe
- Stroke Research in Stoke, Faculty of Medicine & Health Sciences, Keele University, Staffordshire, UK
| | - Danilo Toni
- Neurology Department, University La Sapienza Rome, Rome, Italy
| | - Georgios Tsivgoulis
- Second Department of Neurology, National & Kapodistrian University of Athens, “Attikon” University Hospital, Athens, Greece
| | - Peter Ringleb
- Neurology Department, University Hospital Heidelberg, Heidelberg, Germany
| | - Nils Wahlgren
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Niaz Ahmed
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
39
|
Ko YA, Billheimer JT, Lyssenko NN, Kueider-Paisley A, Wolk DA, Arnold SE, Leung YY, Shaw LM, Trojanowski JQ, Kaddurah-Daouk RF, Kling MA, Rader DJ. ApoJ/Clusterin concentrations are determinants of cerebrospinal fluid cholesterol efflux capacity and reduced levels are associated with Alzheimer's disease. Alzheimers Res Ther 2022; 14:194. [PMID: 36572909 PMCID: PMC9791777 DOI: 10.1186/s13195-022-01119-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 11/06/2022] [Indexed: 12/27/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) shares risk factors with cardiovascular disease (CVD) and dysregulated cholesterol metabolism is a mechanism common to both diseases. Cholesterol efflux capacity (CEC) is an ex vivo metric of plasma high-density lipoprotein (HDL) function and inversely predicts incident CVD independently of other risk factors. Cholesterol pools in the central nervous system (CNS) are largely separate from those in blood, and CNS cholesterol excess may promote neurodegeneration. CEC of cerebrospinal fluid (CSF) may be a useful measure of CNS cholesterol trafficking. We hypothesized that subjects with AD and mild cognitive impairment (MCI) would have reduced CSF CEC compared with Cognitively Normal (CN) and that CSF apolipoproteins apoA-I, apoJ, and apoE might have associations with CSF CEC. METHODS We retrieved CSF and same-day ethylenediaminetetraacetic acid (EDTA) plasma from 108 subjects (40 AD; 18 MCI; and 50 CN) from the Center for Neurodegenerative Disease Research biobank at the Perelman School of Medicine, University of Pennsylvania. For CSF CEC assays, we used N9 mouse microglial cells and SH-SY5Y human neuroblastoma cells, and the corresponding plasma assay used J774 cells. Cells were labeled with [3H]-cholesterol for 24 h, had ABCA1 expression upregulated for 6 h, were exposed to 33 μl of CSF, and then were incubated for 2.5 h. CEC was quantified as percent [3H]-cholesterol counts in medium of total counts medium+cells, normalized to a pool sample. ApoA-I, ApoJ, ApoE, and cholesterol were also measured in CSF. RESULTS We found that CSF CEC was significantly lower in MCI compared with controls and was poorly correlated with plasma CEC. CSF levels of ApoJ/Clusterin were also significantly lower in MCI and were significantly associated with CSF CEC. While CSF ApoA-I was also associated with CSF CEC, CSF ApoE had no association with CSF CEC. CSF CEC is significantly and positively associated with CSF Aβ. Taken together, ApoJ/Clusterin may be an important determinant of CSF CEC, which in turn could mitigate risk of MCI and AD risk by promoting cellular efflux of cholesterol or other lipids. In contrast, CSF ApoE does not appear to play a role in determining CSF CEC.
Collapse
Affiliation(s)
- Yi-An Ko
- grid.25879.310000 0004 1936 8972Division of Translational Medicine and Human Research, Perelman School of Medicine, University of Pennsylvania, 11-125 Smilow Center for Translational Research, 3400 Civic Center Blvd, Philadelphia, PA 19104-5158 USA
| | - Jeffrey T. Billheimer
- grid.25879.310000 0004 1936 8972Division of Translational Medicine and Human Research, Perelman School of Medicine, University of Pennsylvania, 11-125 Smilow Center for Translational Research, 3400 Civic Center Blvd, Philadelphia, PA 19104-5158 USA
| | - Nicholas N. Lyssenko
- grid.264727.20000 0001 2248 3398Alzheimer’s Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140 USA
| | - Alexandra Kueider-Paisley
- grid.26009.3d0000 0004 1936 7961Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC 27708 USA
| | - David A. Wolk
- grid.25879.310000 0004 1936 8972Department of Neurology, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Steven E. Arnold
- grid.38142.3c000000041936754XDepartment of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115 USA
| | - Yuk Yee Leung
- grid.25879.310000 0004 1936 8972Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Leslie M. Shaw
- grid.25879.310000 0004 1936 8972Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - John Q. Trojanowski
- grid.25879.310000 0004 1936 8972Division of Translational Medicine and Human Research, Perelman School of Medicine, University of Pennsylvania, 11-125 Smilow Center for Translational Research, 3400 Civic Center Blvd, Philadelphia, PA 19104-5158 USA
| | - Rima F. Kaddurah-Daouk
- grid.26009.3d0000 0004 1936 7961Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC 27708 USA ,grid.26009.3d0000 0004 1936 7961Duke Institute for Brain Sciences, Duke University, Durham, NC 27708 USA ,grid.26009.3d0000 0004 1936 7961Department of Medicine, Duke University, Durham, NC 27708 USA
| | - Mitchel A. Kling
- grid.262671.60000 0000 8828 4546Department of Geriatrics and Gerontology, New Jersey Institute for Successful Aging, Rowan University School of Osteopathic Medicine, 42 E. Laurel Rd., Suite 1800, Stratford, NJ 08084 USA ,grid.25879.310000 0004 1936 8972Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania USA
| | - Daniel J. Rader
- grid.25879.310000 0004 1936 8972Division of Translational Medicine and Human Research, Perelman School of Medicine, University of Pennsylvania, 11-125 Smilow Center for Translational Research, 3400 Civic Center Blvd, Philadelphia, PA 19104-5158 USA
| |
Collapse
|
40
|
Huang S, Liu X, Liu D, Zhang X, Zhang L, Le W, Zhang Y. Pyrylium-Based Derivatization for Rapid Labeling and Enhanced Detection of Cholesterol in Mass Spectrometry Imaging. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2022; 33:2310-2318. [PMID: 36331251 DOI: 10.1021/jasms.2c00271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Cholesterol in the central nervous system has been increasingly found to be closely related to neurodegenerative diseases. Defects in cholesterol metabolism can cause structural and functional disorders of the central nervous system. The detection of abnormal cholesterol is of great significance for the cognition of physiological and pathological states of organisms, and the spatial distribution of cholesterol can also provide more clues for our understanding of the complex mechanism of disease. Here, we developed a novel pyrylium-based derivatization reagent combined with matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) to visualize cholesterol in biological tissues. A new class of charged hydroxyl derivatization reagents was designed and synthesized, and finally 1-(carboxymethyl)-2,4,6-trimethylpyridinium (CTMP) was screened for tissue derivatization of cholesterol. Different from the shortcomings of traditional hydroxyl labeling methods such as harsh reaction conditions and long reaction time, in our study, we combined the advantages of CTMP itself and the EDCl/HOBt reaction system to achieve instant labeling of cholesterol on tissues through two-step activation. In addition, we also reported changes in cholesterol content in different stages and different brain regions during disease development in SOD1 mutant mouse model. The cholesterol derivatization method we developed provides an efficient way to explore the distribution and spatial metabolic network of cholesterol in biological tissues.
Collapse
Affiliation(s)
- Shuai Huang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, PR China
- University of Chinese Academy of Science, Beijing 100039, PR China
| | - Xinxin Liu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, PR China
| | - Dan Liu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, PR China
| | - Xiaozhe Zhang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, PR China
| | - Lihua Zhang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, PR China
| | - Weidong Le
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian 116021, PR China
| | - Yukui Zhang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, PR China
| |
Collapse
|
41
|
Hartmann H, Ho WY, Chang JC, Ling SC. Cholesterol dyshomeostasis in amyotrophic lateral sclerosis: cause, consequence, or epiphenomenon? FEBS J 2022; 289:7688-7709. [PMID: 34469619 DOI: 10.1111/febs.16175] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/10/2021] [Accepted: 08/31/2021] [Indexed: 01/14/2023]
Abstract
Amyotrophic lateral sclerosis (ALS), the most common adult-onset motor neuron disease, is characterized by the selective degeneration of motor neurons leading to paralysis and eventual death. Multiple pathogenic mechanisms, including systemic dysmetabolism, have been proposed to contribute to ALS. Among them, dyslipidemia, i.e., abnormal level of cholesterol and other lipids in the circulation and central nervous system (CNS), has been reported in ALS patients, but without a consensus. Cholesterol is a constituent of cellular membranes and a precursor of steroid hormones, oxysterols, and bile acids. Consequently, optimal cholesterol levels are essential for health. Due to the blood-brain barrier (BBB), cholesterol cannot move between the CNS and the rest of the body. As such, cholesterol metabolism in the CNS is proposed to operate autonomously. Despite its importance, it remains elusive how cholesterol dyshomeostasis may contribute to ALS. In this review, we aim to describe the current state of cholesterol metabolism research in ALS, identify unresolved issues, and provide potential directions.
Collapse
Affiliation(s)
- Hannelore Hartmann
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Wan Yun Ho
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jer-Cherng Chang
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Shuo-Chien Ling
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Healthy Longevity Translational Research Programme, National University Health System, Singapore, Singapore.,Program in Neuroscience and Behavior Disorders, Duke-NUS Medical School, Singapore, Singapore
| |
Collapse
|
42
|
Amelioration of Age-Related Multiple Neuronal Impairments and Inflammation in High-Fat Diet-Fed Rats: The Prospective Multitargets of Geraniol. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4812993. [PMID: 36304965 PMCID: PMC9596245 DOI: 10.1155/2022/4812993] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 08/31/2022] [Accepted: 09/27/2022] [Indexed: 11/18/2022]
Abstract
Neuroinflammation is documented to alter brain function as a consequence of metabolic changes linked with a high-fat diet (HFD). The primary target of this study is to see how geraniol is effective in manipulating age- and diet-associated multiple toxicity and neuroinflammation in HFD-fed rats. Sixty-four adult male Wistar rats were partitioned into two groups: Group 1 (untreated normal young and aged rats) and Group 2 (HFD-fed young and aged rats) that received HFD for 16 weeks before being orally treated with geraniol or chromax for eight weeks. The results revealed a dropping in proinflammatory cytokines (TNF-α and IL-6) and leptin while boosting adiponectin in geraniol-supplemented rats. The liver, kidney, and lipid profiles were improved in geraniol-HFD-treated groups. HFD-induced brain insulin resistance decreased insulin clearance and insulin-degrading enzyme (IDE) levels significantly after geraniol supplementation. Geraniol suppressed acetylcholinesterase (AChE) activity and alleviated oxidative stress by boosting neuronal reduced glutathione (GSH), catalase (CAT), glutathione-S-transferase (GST), and superoxide dismutase (SOD) activities. It lowered malondialdehyde concentration (TBARS), nitric oxide (NO), and xanthine oxidase (XO) and restored the structural damage to the brain tissue caused by HFD. Compared with model rats, geraniol boosted learning and memory function and ameliorated the inflammation status in the brain by lowering the protein levels of IL-1β, iNOS, NF-κBp65, and COX-2. In addition, the expression levels of inflammation-related genes (MCP-1, TNF-α, IL-6, IL-1β, and IDO-1) were lessened significantly. Remarkably, the supplementation of geraniol reversed the oxidative and inflammation changes associated with aging. It affected the redox status of young rats. In conclusion, our results exhibit the effectiveness of dietary geraniol supplementation in modifying age-related neuroinflammation and oxidative stress in rats and triggering off the use of geraniol as a noninvasive natural compound for controlling age- and diet-associated neuronal impairments and toxicity.
Collapse
|
43
|
Shahid R, Naik SS, Ramphall S, Rijal S, Prakash V, Ekladios H, Mulayamkuzhiyil Saju J, Mandal N, Kham NI, Hamid P. Neurocognitive Impairment in Cardiovascular Disease Patients Taking Statins Versus Proprotein Convertase Subtilisin/Kexin Type 9 (PCSK9) Inhibitors: A Systematic Review. Cureus 2022; 14:e30942. [PMID: 36465767 PMCID: PMC9712061 DOI: 10.7759/cureus.30942] [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: 09/01/2022] [Accepted: 10/31/2022] [Indexed: 11/05/2022] Open
Abstract
Cardiovascular diseases (CVDs) are prevalent medical conditions affecting millions of people worldwide and are associated with significant morbidity and mortality. The main precursor of CVDs and the related events, such as hypertension and heart failure, is hyperlipidemia, most commonly an increase in low-density lipoproteins. Lipid-lowering drugs are cardinal in the treatment of CVDs. American College of Cardiology and American Heart Association have issued guidelines for lipid-lowering therapy, and statins are first-line medication. In the recent years, a new class of lipid-lowering agents called proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors has been identified as the potential lipid-lowering therapy for the statin-resistant patient. In clinical trials and observational studies, PCSK9 inhibitors and statins are both associated with the development of neurocognitive dysfunction in the older population. This systematic review aims to inquire if there is significant neurocognitive dysfunction associated with statins and PCSK9 inhibitors and compare neurocognitive effects associated with statins with those of PCSK9 inhibitors.
Collapse
Affiliation(s)
- Rabia Shahid
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Shaili S Naik
- Internal Medicine, Surat Municipal Institute of Medical Education & Research (SMIMER), Surat, IND
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Shivana Ramphall
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Swarnima Rijal
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Vishakh Prakash
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
- Internal Medicine, Government Medical College, Kozhikode, IND
| | - Heba Ekladios
- Psychiatry, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Jiya Mulayamkuzhiyil Saju
- Internal Medicine, Sree Narayana Institute of Medical Sciences, Ernakulam, IND
- General Surgery, Government Medical College, Thiruvananthapuram, IND
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Naishal Mandal
- Internal Medicine, Surat Municipal Institute of Medical Education & Research (SMIMER), Surat, IND
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Nang I Kham
- Hospital Medicine, University of Medicine 1, Yangon, Yangon, MMR
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Pousette Hamid
- Neurology, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| |
Collapse
|
44
|
Kelley DP, Chaichi A, Duplooy A, Singh D, Gartia MR, Francis J. Labelfree mapping and profiling of altered lipid homeostasis in the rat hippocampus after traumatic stress: Role of oxidative homeostasis. Neurobiol Stress 2022; 20:100476. [PMID: 36032405 PMCID: PMC9403561 DOI: 10.1016/j.ynstr.2022.100476] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 07/27/2022] [Accepted: 07/27/2022] [Indexed: 11/17/2022] Open
Abstract
Oxidative and lipid homeostasis are altered by stress and trauma and post-traumatic stress disorder (PTSD) is associated with alterations to lipid species in plasma. Stress-induced alterations to lipid oxidative and homeostasis may exacerbate PTSD pathology, but few preclinical investigations of stress-induced lipidomic changes in the brain exist. Currently available techniques for the quantification of lipid species in biological samples require tissue extraction and are limited in their ability to retrieve spatial information. Raman imaging can overcome this limitation through the quantification of lipid species in situ in minimally processed tissue slices. Here, we utilized a predator exposure and psychosocial stress (PE/PSS) model of traumatic stress to standardize Raman imaging of lipid species in the hippocampus using LC-MS based lipidomics and these data were confirmed with qRT-PCR measures of mRNA expression of relevant enzymes and transporters. Electron Paramagnetic Resonance Spectroscopy (EPR) was used to measure free radical production and an MDA assay to measure oxidized polyunsaturated fatty acids. We observed that PE/PSS is associated with increased cholesterol, altered lipid concentrations, increased free radical production and reduced oxidized polyunsaturated fats (PUFAs) in the hippocampus (HPC), indicating shifts in lipid and oxidative homeostasis in the HPC after traumatic stress.
Collapse
Affiliation(s)
- D. Parker Kelley
- Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, 70803, USA
| | - Ardalan Chaichi
- Department of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, LA, 70803, USA
| | - Alexander Duplooy
- Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, 70803, USA
| | - Dhirendra Singh
- Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, 70803, USA
| | - Manas Ranjan Gartia
- Department of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, LA, 70803, USA
| | - Joseph Francis
- Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, 70803, USA
| |
Collapse
|
45
|
Borràs C, Mercer A, Sirisi S, Alcolea D, Escolà-Gil JC, Blanco-Vaca F, Tondo M. HDL-like-Mediated Cell Cholesterol Trafficking in the Central Nervous System and Alzheimer's Disease Pathogenesis. Int J Mol Sci 2022; 23:ijms23169356. [PMID: 36012637 PMCID: PMC9409363 DOI: 10.3390/ijms23169356] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/10/2022] [Accepted: 08/18/2022] [Indexed: 01/02/2023] Open
Abstract
The main aim of this work is to review the mechanisms via which high-density lipoprotein (HDL)-mediated cholesterol trafficking through the central nervous system (CNS) occurs in the context of Alzheimer’s disease (AD). Alzheimer’s disease is characterized by the accumulation of extracellular amyloid beta (Aβ) and abnormally hyperphosphorylated intracellular tau filaments in neurons. Cholesterol metabolism has been extensively implicated in the pathogenesis of AD through biological, epidemiological, and genetic studies, with the APOE gene being the most reproducible genetic risk factor for the development of AD. This manuscript explores how HDL-mediated cholesterol is transported in the CNS, with a special emphasis on its relationship to Aβ peptide accumulation and apolipoprotein E (ApoE)-mediated cholesterol transport. Indeed, we reviewed all existing works exploring HDL-like-mediated cholesterol efflux and cholesterol uptake in the context of AD pathogenesis. Existing data seem to point in the direction of decreased cholesterol efflux and the impaired entry of cholesterol into neurons among patients with AD, which could be related to impaired Aβ clearance and tau protein accumulation. However, most of the reviewed studies have been performed in cells that are not physiologically relevant for CNS pathology, representing a major flaw in this field. The ApoE4 genotype seems to be a disruptive element in HDL-like-mediated cholesterol transport through the brain. Overall, further investigations are needed to clarify the role of cholesterol trafficking in AD pathogenesis.
Collapse
Affiliation(s)
- Carla Borràs
- Institut d’Investigació Biomèdica Sant Pau (IIB), Sant Quintí 77-79, 08041 Barcelona, Spain
- CIBERDEM, ISCIII, 28029 Madrid, Spain
- Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Aina Mercer
- Institut d’Investigació Biomèdica Sant Pau (IIB), Sant Quintí 77-79, 08041 Barcelona, Spain
| | - Sònia Sirisi
- Institut d’Investigació Biomèdica Sant Pau (IIB), Sant Quintí 77-79, 08041 Barcelona, Spain
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain
| | - Daniel Alcolea
- Institut d’Investigació Biomèdica Sant Pau (IIB), Sant Quintí 77-79, 08041 Barcelona, Spain
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain
- CIBERNED, ISCIII, 28029 Madrid, Spain
| | - Joan Carles Escolà-Gil
- Institut d’Investigació Biomèdica Sant Pau (IIB), Sant Quintí 77-79, 08041 Barcelona, Spain
- CIBERDEM, ISCIII, 28029 Madrid, Spain
- Correspondence: (J.C.E.-G.); (M.T.); Tel.: +34-93-553-7358 (J.C.E.-G. & M.T.)
| | - Francisco Blanco-Vaca
- Institut d’Investigació Biomèdica Sant Pau (IIB), Sant Quintí 77-79, 08041 Barcelona, Spain
- CIBERDEM, ISCIII, 28029 Madrid, Spain
- Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
- Department of Biochemistry, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain
| | - Mireia Tondo
- Institut d’Investigació Biomèdica Sant Pau (IIB), Sant Quintí 77-79, 08041 Barcelona, Spain
- CIBERDEM, ISCIII, 28029 Madrid, Spain
- Department of Biochemistry, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain
- Correspondence: (J.C.E.-G.); (M.T.); Tel.: +34-93-553-7358 (J.C.E.-G. & M.T.)
| |
Collapse
|
46
|
de Toledo VHC, Feltrin AS, Barbosa AR, Tahira AC, Brentani H. Sex differences in gene regulatory networks during mid-gestational brain development. Front Hum Neurosci 2022; 16:955607. [PMID: 36061507 PMCID: PMC9428411 DOI: 10.3389/fnhum.2022.955607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 07/19/2022] [Indexed: 11/13/2022] Open
Abstract
Neurodevelopmental disorders differ considerably between males and females, and fetal brain development is one of the most critical periods to determine risk for these disorders. Transcriptomic studies comparing male and female fetal brain have demonstrated that the highest difference in gene expression occurs in sex chromosomes, but several autossomal genes also demonstrate a slight difference that has not been yet explored. In order to investigate biological pathways underlying fetal brain sex differences, we applied medicine network principles using integrative methods such as co-expression networks (CEMiTool) and regulatory networks (netZoo). The pattern of gene expression from genes in the same pathway tend to reflect biologically relevant phenomena. In this study, network analysis of fetal brain expression reveals regulatory differences between males and females. Integrating two different bioinformatics tools, our results suggest that biological processes such as cell cycle, cell differentiation, energy metabolism and extracellular matrix organization are consistently sex-biased. MSET analysis demonstrates that these differences are relevant to neurodevelopmental disorders, including autism.
Collapse
Affiliation(s)
- Victor Hugo Calegari de Toledo
- Departamento e Instituto de Psiquiatria, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
- Laboratório de Psicopatologia e Terapêutica Psiquiátrica (LIM23), Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
| | | | | | - Ana Carolina Tahira
- Laboratório de Expressão Gênica, Departamento de Parasitologia, Instituto Butantan, São Paulo, Brazil
| | - Helena Brentani
- Departamento e Instituto de Psiquiatria, Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
- Laboratório de Psicopatologia e Terapêutica Psiquiátrica (LIM23), Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo, Brazil
- *Correspondence: Helena Brentani
| |
Collapse
|
47
|
Sarkar P, Chattopadhyay A. Statin-induced Increase in Actin Polymerization Modulates GPCR Dynamics and Compartmentalization. Biophys J 2022:S0006-3495(22)00708-1. [DOI: 10.1016/j.bpj.2022.08.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/16/2022] [Accepted: 08/25/2022] [Indexed: 11/29/2022] Open
|
48
|
Ho WY, Hartmann H, Ling SC. Central nervous system cholesterol metabolism in health and disease. IUBMB Life 2022; 74:826-841. [PMID: 35836360 DOI: 10.1002/iub.2662] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 06/23/2022] [Indexed: 12/19/2022]
Abstract
Cholesterol is a ubiquitous and essential component of cellular membranes, as it regulates membrane structure and fluidity. Furthermore, cholesterol serves as a precursor for steroid hormones, oxysterol, and bile acids, that are essential for maintaining many of the body's metabolic processes. The biosynthesis and excretion of cholesterol is tightly regulated in order to maintain homeostasis. Although virtually all cells have the capacity to make cholesterol, the liver and brain are the two main organs producing cholesterol in mammals. Once produced, cholesterol is transported in the form of lipoprotein particles to other cell types and tissues. Upon formation of the blood-brain barrier (BBB) during embryonic development, lipoproteins cannot move between the central nervous system (CNS) and the rest of the body. As such, cholesterol biosynthesis and metabolism in the CNS operate autonomously without input from the circulation system in normal physiological conditions. Nevertheless, similar regulatory mechanisms for maintaining cholesterol homeostasis are utilized in both the CNS and peripheral systems. Here, we discuss the functions and metabolism of cholesterol in the CNS. We further focus on how different CNS cell types contribute to cholesterol metabolism, and how ApoE, the major CNS apolipoprotein, is involved in normal and pathophysiological functions. Understanding these basic mechanisms will aid our ability to elucidate how CNS cholesterol dysmetabolism contributes to neurogenerative diseases.
Collapse
Affiliation(s)
- Wan Y Ho
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Program in Neuroscience and Behavior Disorders, Duke-NUS Medical School, Singapore
| | - Hannelore Hartmann
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Shuo-Chien Ling
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Program in Neuroscience and Behavior Disorders, Duke-NUS Medical School, Singapore.,Healthy Longevity Translational Research Programme, National University Health System, Singapore
| |
Collapse
|
49
|
Shapiro LR, Kennedy KG, Dimick MK, Goldstein BI. Elevated atherogenic lipid profile in youth with bipolar disorder during euthymia and hypomanic/mixed but not depressive states. J Psychosom Res 2022; 156:110763. [PMID: 35193092 DOI: 10.1016/j.jpsychores.2022.110763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 02/10/2022] [Accepted: 02/13/2022] [Indexed: 10/19/2022]
Abstract
OBJECTIVE Abnormal blood lipid levels are common in individuals with bipolar disorder (BD). Previous studies have revealed lipid-mood associations in adults with BD, but no data on this relationship is available in youth populations. This cross-sectional study examined the associations of lipid levels with mood states and symptoms in a cohort of youth with BD. METHODS Participants were youth with BD and healthy controls (HCs) between the ages of 13-20 years. We compared the levels of total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), triglycerides (TG), and TG/HDL-C ratio between 4 participant episode groups: BD-euthymic (n = 28), BD-depressed (n = 29) BD-hypomanic/mixed (n = 31), and HCs (n = 89). We also examined for dimensional associations of lipids with mania and depression scores in the overall BD group and within BD episode subgroups. RESULTS TG levels were significantly higher in the BD-euthymic group (p = 0.008, d = 0.59) and in the BD-mixed/hypomanic group (p = 0.03, d = 0.44) compared to the HC group. TG/HDL-C ratio was also higher in the BD-euthymic group compared to the HC group (p = 0.01, d = 0.51). No dimensional associations were found between lipids and mood symptom scores in the overall BD group. However, within the BD-mixed/hypomanic subgroup, higher mania scores were associated with higher TG (β = 0.42, p = 0.04), TG/HDL-C ratio (β = 0.59, p = 0.002), and lower HDL-C (β = 0.56 p = 0.002). CONCLUSIONS Youth with BD demonstrate atherogenic lipid profiles. Higher atherogenic lipids were associated with hypomanic but, contrasting adult BD studies, not depressive symptoms. Future prospective studies are warranted to evaluate the temporal association between lipids and mood among youth with BD.
Collapse
Affiliation(s)
- Lila R Shapiro
- Centre for Youth Bipolar Disorder, Centre for Addictions and Mental Health, Toronto, Canada; Department of Pharmacology, University of Toronto, Toronto, Canada
| | - Kody G Kennedy
- Centre for Youth Bipolar Disorder, Centre for Addictions and Mental Health, Toronto, Canada; Department of Pharmacology, University of Toronto, Toronto, Canada
| | - Mikaela K Dimick
- Centre for Youth Bipolar Disorder, Centre for Addictions and Mental Health, Toronto, Canada; Department of Pharmacology, University of Toronto, Toronto, Canada
| | - Benjamin I Goldstein
- Centre for Youth Bipolar Disorder, Centre for Addictions and Mental Health, Toronto, Canada; Department of Pharmacology, University of Toronto, Toronto, Canada.
| |
Collapse
|
50
|
Lewis MM, Albertson RM, Du G, Kong L, Foy A, Huang X. Parkinson’s Disease Progression and Statins: Hydrophobicity Matters. JOURNAL OF PARKINSON'S DISEASE 2022; 12:821-830. [PMID: 34958045 PMCID: PMC10141621 DOI: 10.3233/jpd-212819] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: Recent randomized clinical trials using hydrophobic statins reported no influence on Parkinson’s disease (PD) clinical progression. Hydrophobicity is a key determinant for blood-brain barrier penetrance. Objective: Investigate a potential effect of statins on PD progression. Methods: Statin use was determined at baseline and subtyped according to hydrophobicity in 125 PD patients participating in the PD Biomarker Program (PDBP, 2012–2015) at our site. Clinical (N = 125) and susceptibility MRI (N = 86) data were obtained at baseline and 18-months. Movement Disorders Society-Unified PD Rating Scales were used to track progression of non-motor (MDS-UPDRS-I) and motor (MDS-UPDRS-II) symptoms, and rater-based scores (MDS-UPDRS-III) of patients in the “on” drug state. R2* values were used to capture pathological progression in the substantia nigra. Associations between statin use, its subtypes, and PD progression were evaluated with linear mixed effect regressions. Results: Compared to statin non-users, overall statin or lipophilic statin use did not significantly influence PD clinical or imaging progression. Hydrophilic statin users, however, demonstrated faster clinical progression of non-motor symptoms [MDS-UPDRS-I (β= 4.8, p = 0.010)] and nigral R2* (β= 3.7, p = 0.043). A similar trend was found for MDS-UPDRS-II (β= 3.9, p = 0.10), but an opposite trend was observed for rater-based MDS-UPDRS-III (β= –7.3, p = 0.10). Compared to lipophilic statin users, hydrophilic statin users also showed significantly faster clinical progression of non-motor symptoms [MDS-UPDRS-I (β= 5.0, p = 0.020)], but R2* did not reach statistical significance (β= 2.5, p = 0.24). Conclusion: This study suggests that hydrophilic, but not lipophilic, statins may be associated with faster PD progression. Future studies may have clinical and scientific implications.
Collapse
Affiliation(s)
- Mechelle M. Lewis
- Department of Neurology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA
- Department of Pharmacology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Richard M. Albertson
- Department of Neurology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Guangwei Du
- Department of Neurology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Lan Kong
- Department of Public Health Sciences, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Andrew Foy
- Department of Public Health Sciences, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA
- Department of Medicine, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Xuemei Huang
- Department of Neurology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA
- Department of Pharmacology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA
- Department of Radiology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA
- Department of Neurosurgery, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA
- Department of Kinesiology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA
| |
Collapse
|