1
|
Zhao S, Jiang J, Zhang J, Jin X. Mediation Mendelian randomization analysis of immune cell phenotypes and glioma risk: unveiling the regulation of cerebrospinal fluid metabolites. Discov Oncol 2025; 16:712. [PMID: 40343558 PMCID: PMC12064550 DOI: 10.1007/s12672-025-02499-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 04/25/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND AND OBJECTIVE Gliomas, particularly glioblastoma multiforme (GBM), are the most common primary central nervous system tumors in adults and are notoriously difficult to treat due to their high heterogeneity and invasiveness. Despite advances in molecular diagnostics and personalized therapies, prognosis remains poor. The immune system plays a critical role in glioma progression. This study employed mediation Mendelian randomization analysis to explore the relationships between immune cell phenotypes, cerebrospinal fluid metabolites, and glioma, aiming to uncover potential mechanisms of tumor progression and immune evasion. METHOD In this study, we employed several analytical methods including IVW, MR Egger, Simple mode, Weighted median, and Weighted mode, with IVW results being considered the primary basis. We assessed heterogeneity and pleiotropy, and used leave-one-out analysis to determine sensitivity, ensuring the stability and reliability of the results. The potential mediating effects of cerebrospinal fluid metabolites were investigated to explore the underlying mechanisms linking immune cell function and glioma. The GWAS data for immune cells, cerebrospinal fluid metabolites, and glioma used in this study were sourced from public databases. RESULT We identified nine risk immune cell phenotypes for glioma (such as CD19 on IgD( +) CD24(-)), and ten protective immune cell phenotypes (such as CD11c on monocytes). Mediation analysis revealed that levels of 7-alpha-hydroxy-3-oxo-4-cholestenoate (7-hoca) (MP = - 14.6%) and Palmitoyl dihydrosphingomyelin (d18:0/16:0) (MP = 7.9%) partially mediated the relationship between CD3 on CD39( +) resting Treg cells and glioma. Additionally, 7-hoca levels (MP = - 12.3%) and Phenyllactate (pla) levels (MP = 4.12%) partially mediated the association between FSC-A on NKT cells and glioma. Furthermore, Glycerophosphoinositol levels (MP = - 12.1%) and Orotate levels (MP = - 11.4%) partially mediated the relationship between Granulocyte adenylyl cyclase (Granulocyte AC) and glioma. CONCLUSION This study identified that specific immune cell phenotypes directly influence glioma risk and indirectly modulate this risk through cerebrospinal fluid metabolites. CD19 on IgD( +) CD24(-) B cells were identified as risk factors, while CD11c on monocytes were protective. Metabolites like 7-hoca and glycerophosphoinositol play key mediating roles. These findings enhance our understanding of glioma pathophysiology and suggest that immune modulation and metabolic intervention may be promising therapeutic strategies.
Collapse
Affiliation(s)
- Siyuan Zhao
- Emergency Center, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuchang, Wuhan, 430071, Hubei, China
- The Second Clinical College, Wuhan University, Wuhan, 430071, Hubei, China
| | - Jinghao Jiang
- Emergency Department, Guilin People's Hospital, 12 Wenming Road, Xiangshan, Guilin, 541000, Guangxi, China
| | - Jianwu Zhang
- Department of Laboratory, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, Hubei, China.
| | - Xiaoqing Jin
- Emergency Center, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuchang, Wuhan, 430071, Hubei, China.
| |
Collapse
|
2
|
An Y, Xi Y, Wang T, Ju M, Feng W, Guo Z, Sun X, Yang K, Qi C, Xiao R. A panel of altered blood oxysterols in patients with mild cognitive impairment: A novel combined diagnostic marker. Pharmacol Res 2025; 213:107661. [PMID: 39984005 DOI: 10.1016/j.phrs.2025.107661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 02/05/2025] [Accepted: 02/14/2025] [Indexed: 02/23/2025]
Abstract
Perturbed cholesterol metabolism may play an important role in the development of dementia and its preclinical stage, mild cognitive impairment (MCI). Oxysterols, the metabolites generated during cholesterol oxidation, also appear to be risk factors for MCI. Therefore, we aimed to investigate if the metabolic profile of blood oxysterols could be used to characterize MCI risk. This cross-sectional study incorporated 501 participants-253 patients with MCI and 248 cognitively normal controls. Serum levels of 22 free oxysterols were measured, and a set of 27 oxysterol-related gene polymorphisms was genotyped. Five [27-hydroxycholesterol (27-OHC), 27-OHC periphery-derived metabolite 3β-hydroxy-5-cholestenoic acid (27-CA) and brain-derived metabolite 7α-hydroxy-3-oxo-4-cholestenoic acid (7-HOCA), 4β-hydroxycholesterol (4β-OHC); 4α-hydroxycholesterol (4α-OHC)] of the twenty-two oxysterols detected in serum significantly differed between the patients with MCI and controls, greatly distinguishing patients with MCI from control individuals (AUC=0.834, 95 % CI: 0.804-0.866). Association analyses demonstrated significant correlations between these candidate oxysterol biomarkers with younger age, higher blood lipids, worse cognitive performance, and higher monounsaturated fatty acid intake. This panel of serum free oxysterols as candidate serum oxysterol biomarkers for MCI highlighted the essential role of 27-OHC in the pathogenesis of early dementia prevention. (The study registered in the Chinese Clinical Trial Registry as ChiCTR-OOC-17011882).
Collapse
Affiliation(s)
- Yu An
- School of Public Health, Capital Medical University, Beijing 100069, China; Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Yuandi Xi
- School of Public Health, Capital Medical University, Beijing 100069, China
| | - Tao Wang
- School of Public Health, Capital Medical University, Beijing 100069, China
| | - Mengwei Ju
- School of Public Health, Capital Medical University, Beijing 100069, China
| | - Wenjing Feng
- School of Public Health, Capital Medical University, Beijing 100069, China
| | - Zhiting Guo
- School of Public Health, Capital Medical University, Beijing 100069, China
| | - Xuejing Sun
- School of Public Health, Capital Medical University, Beijing 100069, China
| | - Kexin Yang
- School of Public Health, Capital Medical University, Beijing 100069, China
| | - Chengyan Qi
- School of Public Health, Capital Medical University, Beijing 100069, China
| | - Rong Xiao
- School of Public Health, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
3
|
Harvey LD, Alotaibi M, Tai YY, Tang Y, Kim HJJ, Kelly NJ, Sun W, Woodcock CSC, Arshad S, Culley MK, El Khoury W, Xie R, Al Aaraj Y, Zhao J, Hafeez N, Rao RJ, Jiang S, Negi V, Kirillova A, Perk D, Watson AM, St. Croix CM, Stolz DB, Lee JY, Cheng MH, Zhang M, Detmer S, Guzman E, Manan RS, Saggar R, Haley KJ, Waxman AB, Okawa S, Schwantes-An TH, Pauciulo MW, Wang B, Webb A, Chauvet C, Anderson DG, Nichols WC, Desai AA, Lafyatis R, Nouraie SM, Wu H, McDonald JG, Cheng S, Bahar I, Bertero T, Benza RL, Jain M, Chan SY. Lysosomal dysfunction and inflammatory sterol metabolism in pulmonary arterial hypertension. Science 2025; 387:eadn7277. [PMID: 39847635 PMCID: PMC12087357 DOI: 10.1126/science.adn7277] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 11/21/2024] [Indexed: 01/25/2025]
Abstract
Vascular inflammation regulates endothelial pathophenotypes, particularly in pulmonary arterial hypertension (PAH). Dysregulated lysosomal activity and cholesterol metabolism activate pathogenic inflammation, but their relevance to PAH is unclear. Nuclear receptor coactivator 7 (NCOA7) deficiency in endothelium produced an oxysterol and bile acid signature through lysosomal dysregulation, promoting endothelial pathophenotypes. This oxysterol signature overlapped with a plasma metabolite signature associated with human PAH mortality. Mice deficient for endothelial Ncoa7 or exposed to an inflammatory bile acid developed worsened PAH. Genetic predisposition to NCOA7 deficiency was driven by single-nucleotide polymorphism rs11154337, which alters endothelial immunoactivation and is associated with human PAH mortality. An NCOA7-activating agent reversed endothelial immunoactivation and rodent PAH. Thus, we established a genetic and metabolic paradigm that links lysosomal biology and oxysterol processes to endothelial inflammation and PAH.
Collapse
Affiliation(s)
- Lloyd D. Harvey
- Medical Scientist Training Program, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mona Alotaibi
- Division of Pulmonary and Critical Care Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA
| | - Yi-Yin Tai
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Ying Tang
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Hee-Jung J. Kim
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Neil J. Kelly
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Division of Cardiology, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA
| | - Wei Sun
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Chen-Shan C. Woodcock
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sanya Arshad
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Miranda K. Culley
- Medical Scientist Training Program, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Wadih El Khoury
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Rong Xie
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Yassmin Al Aaraj
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jingsi Zhao
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Neha Hafeez
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Rashmi J. Rao
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Siyi Jiang
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Vinny Negi
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Anna Kirillova
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Dror Perk
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Annie M. Watson
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | - Donna B. Stolz
- Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ji Young Lee
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY, USA
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA
| | - Mary Hongying Cheng
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY, USA
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA
| | - Manling Zhang
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Division of Cardiology, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA
| | - Samuel Detmer
- Department of Chemistry, Massachusetts Institute of Technology, Boston, MA, USA
| | - Edward Guzman
- Institute of Medical Engineering and Science, Massachusetts Institute of Technology, Boston, MA, USA
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Rajith S. Manan
- Department of Chemical Engineering, Massachusetts Institute of Technology, Boston, MA, USA
| | - Rajan Saggar
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
- Department of Pathology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Kathleen J. Haley
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Aaron B. Waxman
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Satoshi Okawa
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Tae-Hwi Schwantes-An
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis, IN, USA
- Department of Medicine, Indiana University, Indianapolis, IN, USA
| | - Michael W. Pauciulo
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Bing Wang
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Amy Webb
- Department of Biomedical Informatics, Ohio State University, Columbus, OH, USA
| | - Caroline Chauvet
- Université Côte d’Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), Sophia-Antipolis, Valbonne, France
| | - Daniel G. Anderson
- Institute of Medical Engineering and Science, Massachusetts Institute of Technology, Boston, MA, USA
| | - William C. Nichols
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Ankit A. Desai
- Department of Medicine, Indiana University, Indianapolis, IN, USA
| | - Robert Lafyatis
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - S. Mehdi Nouraie
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Haodi Wu
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jeffrey G. McDonald
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Susan Cheng
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ivet Bahar
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY, USA
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA
| | - Thomas Bertero
- Université Côte d’Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), Sophia-Antipolis, Valbonne, France
| | - Raymond L. Benza
- Division of Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mohit Jain
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA
| | - Stephen Y. Chan
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
4
|
Guo Z, Yu H, Yang K, Feng W, Liu M, Wang T, Xiao R. Quantitative Determination of a Series of Oxysterols by an Optimized LC-MS/MS Analysis in Different Tissue Types. Int J Mol Sci 2024; 26:77. [PMID: 39795936 PMCID: PMC11720652 DOI: 10.3390/ijms26010077] [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: 11/13/2024] [Revised: 12/16/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
Oxysterols, as metabolites of cholesterol, play a key role in cholesterol homeostasis, autophagosome formation, and regulation of immune responses. Disorders in oxysterol metabolism are closely related to the pathogenesis of neurodegenerative diseases. To systematically investigate the profound molecular regulatory mechanisms of neurodegenerative diseases, it is necessary to quantify oxysterols and their metabolites in central and peripheral biospecimens simultaneously and accurately. However, there are a lot of unsolved problems with the existing methods, such as the hindrance of applying a single method to different biological specimens or the challenge of simultaneous quantification due to differential groups on the ends of the oxysterol side chains. Herein, according to the physicochemical properties and structure of oxysterols, an optimized liquid chromatography-tandem mass spectrometry method for the quantification of oxysterols was established by optimizing the sample preparation process, chromatographic conditions, mobile phase pH, and solvent selection. Seven oxysterols were detected by this method, including 27-hydroxycholesterol, 7α-hydroxycholesterol, 7α,27-dihydroxycholesterol, 7-dehydrocholesterol, 7α-hydroxy-3-oxo-4-cholestenoic acid, 3-hydroxy-5-cholestenoic acid, and 24(S)-hydroxycholesterol. Non-derivatization extraction with methyl tert-butyl ether was used for different biospecimens, followed by simultaneous chromatographic separation of oxysterols on a phenyl hexyl column. By repeated validation, this method exhibited satisfactory linearity, precision, recovery, sensitivity, repeatability, and stability, and it was successfully applied to the detection of oxysterols in the plasma, cerebral cortex, and liver of mouse. In summary, our optimized method enables concurrent analysis and quantification of oxysterols and their metabolites in various biospecimens, presenting a broad range of applicability.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Rong Xiao
- School of Public Health, Capital Medical University, Beijing 100069, China; (Z.G.); (H.Y.); (K.Y.); (W.F.); (M.L.); (T.W.)
| |
Collapse
|
5
|
Zhao Z, Xing N, Sun G. Identification of 7-HOCA as a Potential Biomarker in Glioblastoma: Evidence from Genome-Wide Association Study and Clinical Validation. Int J Gen Med 2024; 17:6185-6197. [PMID: 39691836 PMCID: PMC11651077 DOI: 10.2147/ijgm.s493488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 11/27/2024] [Indexed: 12/19/2024] Open
Abstract
Purpose Glioblastoma (GBM) is associated with metabolic disturbances, yet the relationships between metabolites with GBM have not been comprehensively explored. This study aims to fill this gap by integrating Mendelian randomization (MR) analysis with clinical validation. Patients and Methods Summary data from genome-wide association study (GWAS) of cerebrospinal fluid (CSF) metabolites, plasma metabolites, and GBM were obtained separately. A total of 338 CSF metabolites and 1400 plasma metabolites were utilized as exposures. Concurrently, GBM was designated as the outcome. A two-sample bidirectional MR study was conducted to investigate the potential association. The inverse variance weighted (IVW) analyses were conducted as causal estimates, accompanied by a series of sensitivity analyses to evaluate the robustness of the results. Additionally, metabolite levels in clinical plasma and CSF samples were quantified using liquid chromatography-mass spectrometry to validate the findings. Results MR analysis identified eight CSF metabolites and six plasma metabolites that were closely associated with GBM. Among these, elevated levels of 7-alpha-hydroxy-3-oxo-4-cholestenoate (7-HOCA) in both CSF and plasma were found to promote GBM. In terms of clinical validation, compared to the control group, 7-HOCA levels were significantly higher in both the CSF and plasma of GBM group. Conclusion This study provides a comprehensive analysis of the metabolic factors contributing to GBM. The identification of specific metabolites, particularly 7-HOCA, that have vital roles in GBM pathogenesis suggests new biomarkers and therapeutic targets, offering potential pathways for improved diagnosis and treatment of GBM.
Collapse
Affiliation(s)
- Zhenxiang Zhao
- Department of Neurosurgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050000, People’s Republic of China
| | - Na Xing
- Department of Endocrinology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050000, People’s Republic of China
| | - Guozhu Sun
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, People’s Republic of China
| |
Collapse
|
6
|
Bao H, Chen Y, Meng Z, Chu Z. The causal relationship between CSF metabolites and GBM: a two-sample mendelian randomization analysis. BMC Cancer 2024; 24:1119. [PMID: 39251963 PMCID: PMC11382389 DOI: 10.1186/s12885-024-12901-7] [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: 03/25/2024] [Accepted: 09/04/2024] [Indexed: 09/11/2024] Open
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is a highly aggressive primary malignant brain tumor characterized by rapid progression, poor prognosis, and high mortality rates. Understanding the relationship between cerebrospinal fluid (CSF) metabolites and GBM is crucial for identifying potential biomarkers and pathways involved in the pathogenesis of this devastating disease. METHODS In this study, Mendelian randomization (MR) analysis was employed to investigate the causal relationship between 338 CSF metabolites and GBM. The data for metabolites were obtained from a genome-wide association study summary dataset based on 291 individuals, and the GBM data was derived from FinnGen included 91 cases and 174,006 controls of European descent. The Inverse Variance Weighted method was utilized to estimate the causal effects. Supplementary comprehensive assessments of causal effects between CSF metabolites and GBM were conducted using MR-Egger regression, Weighted Median, Simple Mode, and Weighted Mode methods. Additionally, tests for heterogeneity and pleiotropy were performed. RESULTS Through MR analysis, a total of 12 identified metabolites and 2 with unknown chemical properties were found to have a causal relationship with GBM. 1-palmitoyl-2-stearoyl-gpc (16:0/18:0), 7-alpha-hydroxy-3-oxo-4-cholestenoate, Alpha-tocopherol, Behenoyl sphingomyelin (d18:1/22:0), Cysteinylglycine, Maleate, Uracil, Valine, X-12,101, X-12,104 and Butyrate (4:0) are associated with an increased risk of GBM. N1-methylinosine, Stachydrine and Succinylcarnitine (c4-dc) are associated with decreased GBM risk. CONCLUSION In conclusion, this study sheds light on the intricate interplay between CSF metabolites and GBM, offering novel perspectives on disease mechanisms and potential treatment avenues. By elucidating the role of CSF metabolites in GBM pathogenesis, this research contributes to the advancement of diagnostic capabilities and targeted therapeutic interventions for this aggressive brain tumor. Further exploration of these findings may lead to improved management strategies and better outcomes for patients with GBM.
Collapse
Affiliation(s)
- Haijun Bao
- Department of Forensic Medicine, First College for Clinical Medicine, Xuzhou Medical University, 84 West Huaihai Rd, Xuzhou, Jiangsu, 221000, China
- Jiangsu Medical Engineering Research Center of Gene Detection, Xuzhou, Jiangsu, China
| | - Yiyang Chen
- Department of Forensic Medicine, First College for Clinical Medicine, Xuzhou Medical University, 84 West Huaihai Rd, Xuzhou, Jiangsu, 221000, China
| | - Zijun Meng
- Department of Forensic Medicine, First College for Clinical Medicine, Xuzhou Medical University, 84 West Huaihai Rd, Xuzhou, Jiangsu, 221000, China
| | - Zheng Chu
- Department of Forensic Medicine, First College for Clinical Medicine, Xuzhou Medical University, 84 West Huaihai Rd, Xuzhou, Jiangsu, 221000, China.
- Jiangsu Medical Engineering Research Center of Gene Detection, Xuzhou, Jiangsu, China.
| |
Collapse
|
7
|
Mao H, Angelini A, Li S, Wang G, Li L, Patterson C, Pi X, Xie L. CRAT links cholesterol metabolism to innate immune responses in the heart. Nat Metab 2023; 5:1382-1394. [PMID: 37443356 PMCID: PMC10685850 DOI: 10.1038/s42255-023-00844-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 06/13/2023] [Indexed: 07/15/2023]
Abstract
Chronic inflammation is associated with increased risk and poor prognosis of heart failure; however, the precise mechanism that provokes sustained inflammation in the failing heart remains elusive. Here we report that depletion of carnitine acetyltransferase (CRAT) promotes cholesterol catabolism through bile acid synthesis pathway in cardiomyocytes. Intracellular accumulation of bile acid or intermediate, 7α-hydroxyl-3-oxo-4-cholestenoic acid, induces mitochondrial DNA stress and triggers cGAS-STING-dependent type I interferon responses. Furthermore, type I interferon responses elicited by CRAT deficiency substantially increase AIM2 expression and AIM2-dependent inflammasome activation. Genetic deletion of cardiomyocyte CRAT in mice of both sexes results in myocardial inflammation and dilated cardiomyopathy, which can be reversed by combined depletion of caspase-1, cGAS or AIM2. Collectively, we identify a mechanism by which cardiac energy metabolism, cholesterol homeostasis and cardiomyocyte-intrinsic innate immune responses are interconnected via a CRAT-mediated bile acid synthesis pathway, which contributes to chronic myocardial inflammation and heart failure progression.
Collapse
Affiliation(s)
- Hua Mao
- Department of Medicine, Section of Cardiovascular Research, Baylor College of Medicine, Houston, TX, USA
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
| | - Aude Angelini
- Department of Medicine, Section of Cardiovascular Research, Baylor College of Medicine, Houston, TX, USA
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
| | - Shengyu Li
- Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX, USA
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Guangyu Wang
- Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX, USA
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Luge Li
- Department of Medicine, Section of Cardiovascular Research, Baylor College of Medicine, Houston, TX, USA
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
| | - Cam Patterson
- University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Xinchun Pi
- Department of Medicine, Section of Cardiovascular Research, Baylor College of Medicine, Houston, TX, USA
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
| | - Liang Xie
- Department of Medicine, Section of Cardiovascular Research, Baylor College of Medicine, Houston, TX, USA.
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
8
|
Bajaj JS, Tandon P, O'Leary JG, Reddy KR, Garcia-Tsao G, Thuluvath P, Lai JC, Subramanian RM, Vargas HE, Wong F, Fagan A, McGeorge S, Thacker LR, Kamath PS. Admission Serum Metabolites and Thyroxine Predict Advanced Hepatic Encephalopathy in a Multicenter Inpatient Cirrhosis Cohort. Clin Gastroenterol Hepatol 2023; 21:1031-1040.e3. [PMID: 35436625 PMCID: PMC11000256 DOI: 10.1016/j.cgh.2022.03.046] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/07/2022] [Accepted: 03/24/2022] [Indexed: 12/18/2022]
Abstract
BACKGROUND & AIMS Grades 3 to 4 hepatic encephalopathy (advanced HE), also termed brain failure, is an organ failure that defines acute-on-chronic liver failure. It is associated with poor outcomes in cirrhosis but cannot be predicted accurately. We aimed to determine the admission metabolomic biomarkers able to predict the development of advanced HE with subsequent validation. METHODS Prospective inpatient cirrhosis cohorts (multicenter and 2-center validation) without brain failure underwent admission serum collection and inpatient follow-up evaluation. Serum metabolomics were analyzed to predict brain failure on random forest analysis and logistic regression. A separate validation cohort also was recruited. RESULTS The multicenter cohort included 602 patients, of whom 144 developed brain failure (105 only brain failure) 3 days after admission. Unadjusted random forest analysis showed that higher admission microbially derived metabolites and lower isoleucine, thyroxine, and lysophospholipids were associated with brain failure development (area under the curve, 0.87 all; 0.90 brain failure only). Logistic regression area under the curve with only clinical variables significantly improved with metabolites (95% CI 0.65-0.75; P = .005). Four metabolites that significantly added to brain failure prediction were low thyroxine and maltose and high methyl-4-hydroxybenzoate sulfate and 3-4 dihydroxy butyrate. Thyroxine alone also significantly added to the model (P = .05). The validation cohort including 81 prospectively enrolled patients, of whom 11 developed brain failure. Admission hospital laboratory thyroxine levels predicted brain failure development despite controlling for clinical variables with high specificity. CONCLUSIONS In a multicenter inpatient cohort, admission serum metabolites, including thyroxine, predicted advanced HE development independent of clinical factors. Admission low local laboratory thyroxine levels were validated as a predictor of advanced HE development in a separate cohort.
Collapse
Affiliation(s)
- Jasmohan S Bajaj
- Department of Medicine, Virginia Commonwealth University, Richmond Veterans Affairs Medical Center, Richmond, Virginia.
| | - Puneeta Tandon
- Department of Medicine, University of Alberta, Edmonton, Canada
| | | | - K Rajender Reddy
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Paul Thuluvath
- Department of Medicine, Mercy Medical Center, Baltimore, Maryland
| | - Jennifer C Lai
- Department of Medicine, University of California San Francisco, San Francisco, California
| | - Ram M Subramanian
- Department of Medicine, Emory University Medical Center, Atlanta, Georgia
| | - Hugo E Vargas
- Department of Medicine, Mayo Clinic Arizona, Phoenix, Arizona
| | - Florence Wong
- Department of Medicine, University of Toronto, Toronto, Canada
| | - Andrew Fagan
- Department of Medicine, Virginia Commonwealth University, Richmond Veterans Affairs Medical Center, Richmond, Virginia
| | - Sara McGeorge
- Department of Medicine, Virginia Commonwealth University, Richmond Veterans Affairs Medical Center, Richmond, Virginia
| | - Leroy R Thacker
- Department of Medicine, Virginia Commonwealth University, Richmond Veterans Affairs Medical Center, Richmond, Virginia
| | - Patrick S Kamath
- Department of Medicine, Mayo Clinic Rochester, Rochester, Minnesota
| |
Collapse
|
9
|
Batra R, Arnold M, Wörheide MA, Allen M, Wang X, Blach C, Levey AI, Seyfried NT, Ertekin-Taner N, Bennett DA, Kastenmüller G, Kaddurah-Daouk RF, Krumsiek J. The landscape of metabolic brain alterations in Alzheimer's disease. Alzheimers Dement 2023; 19:980-998. [PMID: 35829654 PMCID: PMC9837312 DOI: 10.1002/alz.12714] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 05/12/2022] [Accepted: 05/18/2022] [Indexed: 01/18/2023]
Abstract
INTRODUCTION Alzheimer's disease (AD) is accompanied by metabolic alterations both in the periphery and the central nervous system. However, so far, a global view of AD-associated metabolic changes in the brain has been missing. METHODS We metabolically profiled 500 samples from the dorsolateral prefrontal cortex. Metabolite levels were correlated with eight clinical parameters, covering both late-life cognitive performance and AD neuropathology measures. RESULTS We observed widespread metabolic dysregulation associated with AD, spanning 298 metabolites from various AD-relevant pathways. These included alterations to bioenergetics, cholesterol metabolism, neuroinflammation, and metabolic consequences of neurotransmitter ratio imbalances. Our findings further suggest impaired osmoregulation as a potential pathomechanism in AD. Finally, inspecting the interplay of proteinopathies provided evidence that metabolic associations were largely driven by tau pathology rather than amyloid beta pathology. DISCUSSION This work provides a comprehensive reference map of metabolic brain changes in AD that lays the foundation for future mechanistic follow-up studies.
Collapse
Affiliation(s)
- Richa Batra
- Department of Physiology and Biophysics, Institute for
Computational Biomedicine, Englander Institute for Precision Medicine, Weill Cornell
Medicine, New York, NY 10021, USA
| | - Matthias Arnold
- Department of Psychiatry and Behavioral Sciences, Duke
University, Durham, NC, USA
- Institute of Computational Biology, Helmholtz Zentrum
München—German Research Center for Environmental Health, 85764
Neuherberg, Germany
| | - Maria A. Wörheide
- Institute of Computational Biology, Helmholtz Zentrum
München—German Research Center for Environmental Health, 85764
Neuherberg, Germany
| | - Mariet Allen
- Department of Neuroscience, Mayo Clinic Florida,
Jacksonville, FL, USA
| | - Xue Wang
- Department of Health Sciences Research, Mayo Clinic
Florida, Jacksonville, FL, USA
| | - Colette Blach
- Department of Psychiatry and Behavioral Sciences, Duke
University, Durham, NC, USA
| | - Allan I. Levey
- Goizueta Alzheimer’s Disease Research Center, Emory
University, Atlanta, GA, USA
| | | | - Nilüfer Ertekin-Taner
- Department of Neuroscience, Mayo Clinic Florida,
Jacksonville, FL, USA
- Department of Neurology, Mayo Clinic Florida, Jacksonville,
FL, USA
| | - David A. Bennett
- Rush Alzheimer’s Disease Center, Rush University
Medical Center, Chicago, IL, USA
| | - Gabi Kastenmüller
- Institute of Computational Biology, Helmholtz Zentrum
München—German Research Center for Environmental Health, 85764
Neuherberg, Germany
| | - Rima F. Kaddurah-Daouk
- Department of Psychiatry and Behavioral Sciences, Duke
Institute for Brain Sciences and Department of Medicine, Duke University, Durham,
NC, 27708, USA
| | - Jan Krumsiek
- Department of Physiology and Biophysics, Institute for
Computational Biomedicine, Englander Institute for Precision Medicine, Weill Cornell
Medicine, New York, NY 10021, USA
| |
Collapse
|
10
|
Turk BR, Poisson LM, Nemeth CL, Goodman J, Moser AB, Jones RO, Fatemi A, Singh J. MicroRNA and metabolomics signatures for adrenomyeloneuropathy disease severity. JIMD Rep 2022; 63:593-603. [PMID: 36341174 PMCID: PMC9626672 DOI: 10.1002/jmd2.12323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 07/14/2022] [Accepted: 08/03/2022] [Indexed: 10/04/2023] Open
Abstract
Adrenomyeloneuropathy (AMN), the slow progressive phenotype of adrenoleukodystrophy (ALD), has no clinical plasma biomarker for disease progression. This feasibility study aimed to determine whether metabolomics and micro-RNA in blood plasma provide a potential source of biomarkers for AMN disease severity. Metabolomics and RNA-seq were performed on AMN and healthy human blood plasma. Biomarker discovery and pathway analyses were performed using clustering, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis, and regression against patient's clinical Expanded Disability Status Score (EDSS). Fourteen AMN and six healthy control samples were analyzed. AMN showed strong disease-severity-specific metabolic and miRNA clustering signatures. Strong, significant clinical correlations were shown for 7-alpha-hydroxy-3-oxo-4-cholestenoate (7-HOCA) (r 2 = 0.83, p < 0.00001), dehydroepiandrosterone sulfate (DHEA-S; r 2 = 0.82, p < 0.00001), hypoxanthine (r 2 = 0.82, p < 0.00001), as well as miRNA-432-5p (r 2 = 0.68, p < 0.00001). KEGG pathway comparison of mild versus severe disease identified affected downstream systems: GAREM, IGF-1, CALCRL, SMAD2&3, glutathione peroxidase, LDH, and NOS. This feasibility study demonstrates that miRNA and metabolomics are a source of potential plasma biomarkers for disease severity in AMN, providing both a disease signature and individual markers with strong clinical correlations. Network analyses of affected systems implicate differentially altered vascular, inflammatory, and oxidative stress pathways, suggesting disease-severity-specific mechanisms as a function of disease severity.
Collapse
Affiliation(s)
- Bela Rui Turk
- Moser Center for Leukodystrophies, Kennedy Krieger InstituteJohns Hopkins Medical InstitutionsBaltimoreMarylandUSA
| | - Laila Marie Poisson
- Department of Public Health SciencesHenry Ford Health SystemDetroitMichiganUSA
| | - Christina Linnea Nemeth
- Moser Center for Leukodystrophies, Kennedy Krieger InstituteJohns Hopkins Medical InstitutionsBaltimoreMarylandUSA
| | - Jordan Goodman
- Moser Center for Leukodystrophies, Kennedy Krieger InstituteJohns Hopkins Medical InstitutionsBaltimoreMarylandUSA
| | - Ann B. Moser
- Moser Center for Leukodystrophies, Kennedy Krieger InstituteJohns Hopkins Medical InstitutionsBaltimoreMarylandUSA
| | - Richard Owen Jones
- Moser Center for Leukodystrophies, Kennedy Krieger InstituteJohns Hopkins Medical InstitutionsBaltimoreMarylandUSA
| | - Ali Fatemi
- Moser Center for Leukodystrophies, Kennedy Krieger InstituteJohns Hopkins Medical InstitutionsBaltimoreMarylandUSA
| | - Jaspreet Singh
- Department of NeurologyHenry Ford Health SystemDetroitMichiganUSA
| |
Collapse
|
11
|
Ventriculoperitoneal Shunt Treatment Increases 7 Alpha Hy-Droxy-3-Oxo-4-Cholestenoic Acid and 24-Hydroxycholesterol Concentrations in Idiopathic Normal Pressure Hydrocephalus. Brain Sci 2022; 12:brainsci12111450. [DOI: 10.3390/brainsci12111450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 10/26/2022] [Accepted: 10/26/2022] [Indexed: 11/16/2022] Open
Abstract
Idiopathic normal pressure hydrocephalus (iNPH) is the most common form of hydrocephalus in the adult population, and is often treated with cerebrospinal fluid (CSF) drainage using a ventriculoperitoneal (VP) shunt. Symptoms of iNPH include gait impairment, cognitive decline, and urinary incontinence. The pathophysiology behind the symptoms of iNPH is still unknown, and no reliable biomarkers have been established to date. The aim of this study was to investigate the possible use of the oxysterols as biomarkers in this disease. CSF levels of the oxysterols 24S- and 27-hydroxycholesterol, as well as the major metabolite of 27-hydroxycholesterol, 7 alpha hydroxy-3-oxo-4-cholestenoic acid (7HOCA), were measured in iNPH-patients before and after treatment with a VP-shunt. Corresponding measurements were also performed in healthy controls. VP-shunt treatment significantly increased the levels of 7HOCA and 24S-hydroxycholesterol in CSF (p = 0.014 and p = 0.037, respectively). The results are discussed in relation to the beneficial effects of VP-shunt treatment. Furthermore, the possibility that CSF drainage may reduce an inhibitory effect of transiently increased pressure on the metabolic capacity of neuronal cells in the brain is discussed. This capacity includes the elimination of cholesterol by the 24S-hydroxylase mechanisms.
Collapse
|
12
|
Regulation of Th17/Treg Balance by 27-Hydroxycholesterol and 24S-Hydroxycholesterol Correlates with Learning and Memory Ability in Mice. Int J Mol Sci 2022; 23:ijms23084370. [PMID: 35457188 PMCID: PMC9028251 DOI: 10.3390/ijms23084370] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/07/2022] [Accepted: 04/07/2022] [Indexed: 02/07/2023] Open
Abstract
Dysregulation of cholesterol metabolism and its oxidative products-oxysterols-in the brain is known to be associated with neurodegenerative diseases. It is well-known that 27-hydroxycholesterol (27-OHC) and 24S-hydroxycholesterol (24S-OHC) are the main oxysterols contributing to the pathogenesis of Alzheimer's disease (AD). However, the molecular mechanism of how 27-OHC and 24S-OHC cause cognitive decline remains unclear. To verify whether 27-OHC and 24S-OHC affect learning and memory by regulating immune responses, C57BL/6J mice were subcutaneously injected with saline, 27-OHC, 24S-OHC, 27-OHC+24S-OHC for 21 days. The oxysterols level and expression level of related metabolic enzymes, as well as the immunomodulatory factors were measured. Our results indicated that 27-OHC-treated mice showed worse learning and memory ability and higher immune responses, but lower expression level of interleukin-10 (IL-10) and interferon (IFN-λ2) compared with saline-treated mice, while 24S-OHC mice performed better in the Morris water maze test than control mice. No obvious morphological lesion was observed in these 24S-OHC-treated mice. Moreover, the expression level of interleukin-17A (IL-17A), granulocyte-macrophage colony-stimulating factor (GM-CSF) and macrophage inflammatory protein 3α (MIP-3α) were significantly decreased after 24S-OHC treatment. Notably, compared with 27-OHC group, mice treated with 27-OHC+24S-OHC showed higher brain 24S-OHC level, accompanied by increased CYP46A1 expression level while decreased CYP7B1, retinoic acid-related orphan receptor gamma t (RORγt) and IL-17A expression level. In conclusion, our study indicated that 27-OHC is involved in regulating the expression of RORγt, disturbing Th17/Treg balance-related immune responses which may be associated with the learning and memory impairment in mice. In contrast, 24S-OHC is neuroprotective and attenuates the neurotoxicity of 27-OHC.
Collapse
|
13
|
Hysi PG, Mangino M, Christofidou P, Falchi M, Karoly ED, NIHR Bioresource Investigators, Mohney RP, Valdes AM, Spector TD, Menni C. Metabolome Genome-Wide Association Study Identifies 74 Novel Genomic Regions Influencing Plasma Metabolites Levels. Metabolites 2022; 12:61. [PMID: 35050183 PMCID: PMC8777659 DOI: 10.3390/metabo12010061] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/14/2021] [Accepted: 12/16/2021] [Indexed: 01/27/2023] Open
Abstract
Metabolites are small products of metabolism that provide a snapshot of the wellbeing of an organism and the mechanisms that control key physiological processes involved in health and disease. Here we report the results of a genome-wide association study of 722 circulating metabolite levels in 8809 subjects of European origin, providing both breadth and depth. These analyses identified 202 unique genomic regions whose variations are associated with the circulating levels of 478 different metabolites. Replication with a subset of 208 metabolites that were available in an independent dataset for a cohort of 1768 European subjects confirmed the robust associations, including 74 novel genomic regions not associated with any metabolites in previous works. This study enhances our knowledge of genetic mechanisms controlling human metabolism. Our findings have major potential for identifying novel targets and developing new therapeutic strategies.
Collapse
Affiliation(s)
- Pirro G. Hysi
- Department of Twin Research and Genetic Epidemiology, King’s College London, London SE1 7EH, UK; (P.G.H.); (M.M.); (P.C.); (M.F.); (A.M.V.)
| | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology, King’s College London, London SE1 7EH, UK; (P.G.H.); (M.M.); (P.C.); (M.F.); (A.M.V.)
- NIHR Biomedical Research Centre at Guy’s and St. Thomas’ Foundation Trust, London SE1 9RT, UK
| | - Paraskevi Christofidou
- Department of Twin Research and Genetic Epidemiology, King’s College London, London SE1 7EH, UK; (P.G.H.); (M.M.); (P.C.); (M.F.); (A.M.V.)
| | - Mario Falchi
- Department of Twin Research and Genetic Epidemiology, King’s College London, London SE1 7EH, UK; (P.G.H.); (M.M.); (P.C.); (M.F.); (A.M.V.)
| | - Edward D. Karoly
- Discovery and Translational Sciences, Metabolon Inc., Raleigh-Durham, NC 27560, USA; (E.D.K.); (R.P.M.)
| | | | - Robert P. Mohney
- Discovery and Translational Sciences, Metabolon Inc., Raleigh-Durham, NC 27560, USA; (E.D.K.); (R.P.M.)
| | - Ana M. Valdes
- Department of Twin Research and Genetic Epidemiology, King’s College London, London SE1 7EH, UK; (P.G.H.); (M.M.); (P.C.); (M.F.); (A.M.V.)
- Inflammation, Injury and Recovery Sciences, School of Medicine, University of Nottingham, Nottingham NG5 1PB, UK
| | - Tim D. Spector
- Department of Twin Research and Genetic Epidemiology, King’s College London, London SE1 7EH, UK; (P.G.H.); (M.M.); (P.C.); (M.F.); (A.M.V.)
| | - Cristina Menni
- Department of Twin Research and Genetic Epidemiology, King’s College London, London SE1 7EH, UK; (P.G.H.); (M.M.); (P.C.); (M.F.); (A.M.V.)
| |
Collapse
|
14
|
Junker J, Kamp F, Winkler E, Steiner H, Bracher F, Müller C. Effective sample preparation procedure for the analysis of free neutral steroids, free steroid acids and sterol sulfates in different tissues by GC-MS. J Steroid Biochem Mol Biol 2021; 211:105880. [PMID: 33757894 DOI: 10.1016/j.jsbmb.2021.105880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 03/11/2021] [Accepted: 03/11/2021] [Indexed: 10/21/2022]
Abstract
Steroids play an important role in cell regulation and homeostasis. Many diseases like Alzheimer's disease or Smith-Lemli-Opitz syndrome are known to be associated with deviations in the steroid profile. Most published methods only allow the analysis of small subgroups of steroids and cannot give an overview of the total steroid profile. We developed and validated a method that allows the analysis of free neutral steroids, including intermediates of cholesterol biosynthesis, free oxysterols, C19 and C21 steroids, free steroid acids, including bile acids, and sterol sulfates using gas chromatography-mass spectrometry. Samples were analyzed in scan mode for screening purposes and in dynamic multiple reaction monitoring mode for highly sensitive quantitative analysis. The method was validated for mouse brain and liver tissue and consists of sample homogenization, lipid extraction, steroid group separation, deconjugation, derivatization and gas chromatography-mass spectrometry analysis. We applied the method on brain and liver samples of mice (10 months and 3 weeks old) and cultured N2a cells and report the endogenous concentrations of 29 physiological steroids.
Collapse
Affiliation(s)
- Julia Junker
- Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians University-Munich, Butenandtstraße 5-13, 81377, Munich, Germany
| | - Frits Kamp
- Biomedical Center (BMC), Metabolic Biochemistry, Ludwig-Maximilians University-Munich, Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Edith Winkler
- Biomedical Center (BMC), Metabolic Biochemistry, Ludwig-Maximilians University-Munich, Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Harald Steiner
- Biomedical Center (BMC), Metabolic Biochemistry, Ludwig-Maximilians University-Munich, Feodor-Lynen-Straße 17, 81377, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Franz Bracher
- Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians University-Munich, Butenandtstraße 5-13, 81377, Munich, Germany
| | - Christoph Müller
- Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians University-Munich, Butenandtstraße 5-13, 81377, Munich, Germany.
| |
Collapse
|
15
|
Parrado-Fernandez C, Leoni V, Saeed A, Rodriguez-Rodriguez P, Sandebring-Matton A, Córdoba-Beldad CM, Bueno P, Gali CC, Panzenboeck U, Cedazo-Minguez A, Björkhem I. Sex difference in flux of 27-hydroxycholesterol into the brain. Br J Pharmacol 2021; 178:3194-3204. [PMID: 33345295 PMCID: PMC8359195 DOI: 10.1111/bph.15353] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 11/30/2020] [Accepted: 12/05/2020] [Indexed: 01/21/2023] Open
Abstract
Background and Purpose The cerebrospinal fluid (CSF)/plasma albumin ratio (QAlb) is believed to reflect the integrity of the blood–brain barrier (BBB). Recently, we reported that QAlb is lower in females. This may be important for uptake of neurotoxic 27‐hydroxycholesterol (27OH) by the brain in particular because plasma levels of 27OH are higher in males. We studied sex differences in the relation between CSF and plasma levels of 27OH and its major metabolite 7α‐hydroxy‐3‐oxo‐4‐cholestenoic acid (7HOCA) with QAlb. We tested the possibility of sex differences in the brain metabolism of 27OH and if its flux into the brain disrupted integrity of the BBB. Experimental Approach We have examined our earlier studies looking for sex differences in CSF levels of oxysterols and their relation to QAlb. We utilized an in vitro model for the BBB with primary cultured brain endothelial cells to test if 27OH has a disruptive effect on this barrier. We measured mRNA and protein levels of CYP7B1 in autopsy brain samples. Key Results The correlation between CSF levels of 27OH and QAlb was higher in males whereas, with 7HOCA, the correlation was higher in females. No significant sex difference in the expression of CYP7B1 mRNA in brain autopsy samples. A correlation was found between plasma levels of 27OH and QAlb. No support was obtained for the hypothesis that plasma levels of 27OH have a disruptive effect on the BBB. Conclusions and Implications The sex differences are discussed in relation to negative effects of 27OH on different brain functions. LINKED ARTICLES This article is part of a themed issue on Oxysterols, Lifelong Health and Therapeutics. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.16/issuetoc
Collapse
Affiliation(s)
- Cristina Parrado-Fernandez
- Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Solna, Sweden.,Discovery and Research, AlzeCure Pharma AB, Huddinge, Sweden
| | - Valerio Leoni
- Laboratory of Clinical Chemistry, Hospital of Desio, ASST-Monza and School of Medicine, University of Milano Bicocca, Monza, Italy
| | - Ahmed Saeed
- Department of Laboratory Medicine, Karolinska Institute, Huddinge, Sweden
| | | | - Anna Sandebring-Matton
- Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Solna, Sweden
| | | | - Paula Bueno
- Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Solna, Sweden
| | - Chaitanya Chakravarthi Gali
- Division of Immunology and Pathophysiology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Ute Panzenboeck
- Division of Immunology and Pathophysiology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Angel Cedazo-Minguez
- Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Solna, Sweden.,Rare & Neurologic Diseases Research Therapeutic Area, Neurodegeneration Research, Sanofi Pharmaceuticals, Paris, France
| | - Ingemar Björkhem
- Department of Laboratory Medicine, Karolinska Institute, Huddinge, Sweden
| |
Collapse
|
16
|
Yutuc E, Dickson AL, Pacciarini M, Griffiths L, Baker PRS, Connell L, Öhman A, Forsgren L, Trupp M, Vilarinho S, Khalil Y, Clayton PT, Sari S, Dalgic B, Höflinger P, Schöls L, Griffiths WJ, Wang Y. Deep mining of oxysterols and cholestenoic acids in human plasma and cerebrospinal fluid: Quantification using isotope dilution mass spectrometry. Anal Chim Acta 2021; 1154:338259. [PMID: 33736801 PMCID: PMC7988461 DOI: 10.1016/j.aca.2021.338259] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 01/25/2021] [Indexed: 01/01/2023]
Abstract
Both plasma and cerebrospinal fluid (CSF) are rich in cholesterol and its metabolites. Here we describe in detail a methodology for the identification and quantification of multiple sterols including oxysterols and sterol-acids found in these fluids. The method is translatable to any laboratory with access to liquid chromatography – tandem mass spectrometry. The method exploits isotope-dilution mass spectrometry for absolute quantification of target metabolites. The method is applicable for semi-quantification of other sterols for which isotope labelled surrogates are not available and approximate quantification of partially identified sterols. Values are reported for non-esterified sterols in the absence of saponification and total sterols following saponification. In this way absolute quantification data is reported for 17 sterols in the NIST SRM 1950 plasma along with semi-quantitative data for 8 additional sterols and approximate quantification for one further sterol. In a pooled (CSF) sample used for internal quality control, absolute quantification was performed on 10 sterols, semi-quantification on 9 sterols and approximate quantification on a further three partially identified sterols. The value of the method is illustrated by confirming the sterol phenotype of a patient suffering from ACOX2 deficiency, a rare disorder of bile acid biosynthesis, and in a plasma sample from a patient suffering from cerebrotendinous xanthomatosis, where cholesterol 27-hydroxylase is deficient. Absolute quantification of oxysterols and cholestenoic acids. Methodology applicable to plasma and cerebrospinal fluid. Data generated for non-esterified and total sterols. Diastereoisomers at C-24 and C-25 separated and quantified.
Collapse
Affiliation(s)
- Eylan Yutuc
- Swansea University Medical School, ILS1 Building, Singleton Park, Swansea, SA2 8PP, Wales, UK
| | - Alison L Dickson
- Swansea University Medical School, ILS1 Building, Singleton Park, Swansea, SA2 8PP, Wales, UK
| | - Manuela Pacciarini
- Swansea University Medical School, ILS1 Building, Singleton Park, Swansea, SA2 8PP, Wales, UK
| | - Lauren Griffiths
- Swansea University Medical School, ILS1 Building, Singleton Park, Swansea, SA2 8PP, Wales, UK
| | | | | | - Anders Öhman
- Department of Integrative Medical Biology, Umeå University, SE-901 87, Umeå, Sweden
| | - Lars Forsgren
- Department of Clinical Science, Neurosciences, Umeå University, SE-901 85, Umeå, Sweden
| | - Miles Trupp
- Department of Clinical Science, Neurosciences, Umeå University, SE-901 85, Umeå, Sweden
| | - Sílvia Vilarinho
- Departments of Internal Medicine, Section of Digestive Diseases, and of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Youssef Khalil
- Inborn Errors of Metabolism, Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
| | - Peter T Clayton
- Inborn Errors of Metabolism, Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
| | - Sinan Sari
- Department of Pediatrics, Division of Gastroenterology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Buket Dalgic
- Department of Pediatrics, Division of Gastroenterology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Philip Höflinger
- Department of Neurology and Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Ludger Schöls
- Department of Neurology and Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany; German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - William J Griffiths
- Swansea University Medical School, ILS1 Building, Singleton Park, Swansea, SA2 8PP, Wales, UK.
| | - Yuqin Wang
- Swansea University Medical School, ILS1 Building, Singleton Park, Swansea, SA2 8PP, Wales, UK
| |
Collapse
|
17
|
Doerfler H, Botesteanu DA, Blech S, Laux R. Untargeted Metabolomic Analysis Combined With Multivariate Statistics Reveal Distinct Metabolic Changes in GPR40 Agonist-Treated Animals Related to Bile Acid Metabolism. Front Mol Biosci 2021; 7:598369. [PMID: 33521051 PMCID: PMC7843463 DOI: 10.3389/fmolb.2020.598369] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 11/23/2020] [Indexed: 12/11/2022] Open
Abstract
Metabolomics has been increasingly applied to biomarker discovery, as untargeted metabolic profiling represents a powerful exploratory tool for identifying causal links between biomarkers and disease phenotypes. In the present work, we used untargeted metabolomics to investigate plasma specimens of rats, dogs, and mice treated with small-molecule drugs designed for improved glycemic control of type 2 diabetes mellitus patients via activation of GPR40. The in vivo pharmacology of GPR40 is not yet fully understood. Compounds targeting this receptor have been found to induce drug-induced liver injury (DILI). Metabolomic analysis facilitating an integrated UPLC-TWIMS-HRMS platform was used to detect metabolic differences between treated and non-treated animals within two 4-week toxicity studies in rat and dog, and one 2-week toxicity study in mouse. Multivariate statistics of untargeted metabolomics data subsequently revealed the presence of several significantly upregulated endogenous compounds in the treated animals whose plasma level is known to be affected during DILI. A specific bile acid metabolite useful as endogenous probe for drug-drug interaction studies was identified (chenodeoxycholic acid-24 glucuronide), as well as a metabolic precursor indicative of acidic bile acid biosynthesis (7α-hydroxy-3-oxo-4-cholestenoic acid). These results correlate with typical liver toxicity parameters on the individual level.
Collapse
Affiliation(s)
- Hannes Doerfler
- Department of Drug Metabolism & Pharmacokinetics, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Dana-Adriana Botesteanu
- Department of Drug Discovery Sciences, Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - Stefan Blech
- Department of Drug Metabolism & Pharmacokinetics, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Ralf Laux
- Department of Drug Metabolism & Pharmacokinetics, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| |
Collapse
|
18
|
Jimenez-Luna C, Martin-Blazquez A, Dieguez-Castillo C, Diaz C, Martin-Ruiz JL, Genilloud O, Vicente F, del Palacio JP, Prados J, Caba O. Novel Biomarkers to Distinguish between Type 3c and Type 2 Diabetes Mellitus by Untargeted Metabolomics. Metabolites 2020; 10:423. [PMID: 33105675 PMCID: PMC7690399 DOI: 10.3390/metabo10110423] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/16/2020] [Accepted: 10/20/2020] [Indexed: 01/05/2023] Open
Abstract
Pancreatogenic diabetes mellitus (T3cDM) is a highly frequent complication of pancreatic disease, especially chronic pancreatitis, and it is often misdiagnosed as type 2 diabetes mellitus (T2DM). A correct diagnosis allows the appropriate treatment of these patients, improving their quality of life, and various technologies have been employed over recent years to search for specific biomarkers of each disease. The main aim of this metabolomic project was to find differential metabolites between T3cDM and T2DM. Reverse-phase liquid chromatography coupled to high-resolution mass spectrometry was performed in serum samples from patients with T3cDM and T2DM. Multivariate Principal Component and Partial Least Squares-Discriminant analyses were employed to evaluate between-group variations. Univariate and multivariate analyses were used to identify potential candidates and the area under the receiver-operating characteristic (ROC) curve was calculated to evaluate their diagnostic value. A panel of five differential metabolites obtained an area under the ROC curve of 0.946. In this study, we demonstrate the usefulness of untargeted metabolomics for the differential diagnosis between T3cDM and T2DM and propose a panel of five metabolites that appear altered in the comparison between patients with these diseases.
Collapse
Affiliation(s)
- Cristina Jimenez-Luna
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18012 Granada, Spain; (C.J.-L.); (J.P.); (O.C.)
| | - Ariadna Martin-Blazquez
- Fundación MEDINA, Centro de Excelencia para la Investigación en Medicamentos Innovadores en Andalucía, 18012 Granada, Spain; (A.M.-B.); (C.D.); (O.G.); (F.V.)
| | - Carmelo Dieguez-Castillo
- Department of Gastroenterology, San Cecilio University Hospital, 18012 Granada, Spain; (C.D.-C.), (J.L.M.-R.)
| | - Caridad Diaz
- Fundación MEDINA, Centro de Excelencia para la Investigación en Medicamentos Innovadores en Andalucía, 18012 Granada, Spain; (A.M.-B.); (C.D.); (O.G.); (F.V.)
| | - Jose Luis Martin-Ruiz
- Department of Gastroenterology, San Cecilio University Hospital, 18012 Granada, Spain; (C.D.-C.), (J.L.M.-R.)
| | - Olga Genilloud
- Fundación MEDINA, Centro de Excelencia para la Investigación en Medicamentos Innovadores en Andalucía, 18012 Granada, Spain; (A.M.-B.); (C.D.); (O.G.); (F.V.)
| | - Francisca Vicente
- Fundación MEDINA, Centro de Excelencia para la Investigación en Medicamentos Innovadores en Andalucía, 18012 Granada, Spain; (A.M.-B.); (C.D.); (O.G.); (F.V.)
| | - Jose Perez del Palacio
- Fundación MEDINA, Centro de Excelencia para la Investigación en Medicamentos Innovadores en Andalucía, 18012 Granada, Spain; (A.M.-B.); (C.D.); (O.G.); (F.V.)
| | - Jose Prados
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18012 Granada, Spain; (C.J.-L.); (J.P.); (O.C.)
| | - Octavio Caba
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18012 Granada, Spain; (C.J.-L.); (J.P.); (O.C.)
| |
Collapse
|
19
|
Shiffka SJ, Jones JW, Li L, Farese AM, MacVittie TJ, Wang H, Swaan PW, Kane MA. Quantification of common and planar bile acids in tissues and cultured cells. J Lipid Res 2020; 61:1524-1535. [PMID: 32718973 DOI: 10.1194/jlr.d120000726] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Bile acids (BAs) have been established as ubiquitous regulatory molecules implicated in a large variety of healthy and pathological processes. However, the scope of BA heterogeneity is often underrepresented in current literature. This is due in part to inadequate detection methods, which fail to distinguish the individual constituents of the BA pool. Thus, the primary aim of this study was to develop a method that would allow the simultaneous analysis of specific C24 BA species, and to apply that method to biological systems of interest. Herein, we describe the generation and validation of an LC-MS/MS assay for quantification of numerous BAs in a variety of cell systems and relevant biofluids and tissue. These studies included the first baseline level assessment for planar BAs, including allocholic acid, in cell lines, biofluids, and tissue in a nonhuman primate (NHP) laboratory animal, Macaca mulatta, in healthy conditions. These results indicate that immortalized cell lines make poor models for the study of BA synthesis and metabolism, whereas human primary hepatocytes represent a promising alternative model system. We also characterized the BA pool of M. mulatta in detail. Our results support the use of NHP models for the study of BA metabolism and pathology in lieu of murine models. Moreover, the method developed here can be applied to the study of common and planar C24 BA species in other systems.
Collapse
Affiliation(s)
- Stephanie J Shiffka
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland Baltimore, Baltimore, MD, USA
| | - Jace W Jones
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland Baltimore, Baltimore, MD, USA
| | - Linhao Li
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland Baltimore, Baltimore, MD, USA
| | - Ann M Farese
- Department of Radiation Oncology, Division of Translational Radiation Science, School of Medicine, University of Maryland Baltimore, Baltimore, MD, USA
| | - Thomas J MacVittie
- Department of Radiation Oncology, Division of Translational Radiation Science, School of Medicine, University of Maryland Baltimore, Baltimore, MD, USA
| | - Hongbing Wang
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland Baltimore, Baltimore, MD, USA
| | - Peter W Swaan
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland Baltimore, Baltimore, MD, USA
| | - Maureen A Kane
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland Baltimore, Baltimore, MD, USA
| |
Collapse
|
20
|
Localization of sterols and oxysterols in mouse brain reveals distinct spatial cholesterol metabolism. Proc Natl Acad Sci U S A 2020; 117:5749-5760. [PMID: 32132201 PMCID: PMC7084107 DOI: 10.1073/pnas.1917421117] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The brain is a remarkably complex organ and cholesterol homeostasis underpins brain function. It is known that cholesterol is not evenly distributed across different brain regions; however, the precise map of cholesterol metabolism in the brain remains unclear. If cholesterol metabolism is to be correlated with brain function it is essential to generate such a map. Here we describe an advanced mass spectrometry platform to reveal spatial cholesterol metabolism in situ at 400-µm spot diameter on 10-µm tissue slices from mouse brain. We mapped, not only cholesterol, but also other biologically active sterols arising from cholesterol turnover in both wild type and mice lacking cholesterol 24S-hydroxylase (CYP46A1), the major cholesterol metabolizing enzyme. Dysregulated cholesterol metabolism is implicated in a number of neurological disorders. Many sterols, including cholesterol and its precursors and metabolites, are biologically active and important for proper brain function. However, spatial cholesterol metabolism in brain and the resulting sterol distributions are poorly defined. To better understand cholesterol metabolism in situ across the complex functional regions of brain, we have developed on-tissue enzyme-assisted derivatization in combination with microliquid extraction for surface analysis and liquid chromatography-mass spectrometry to locate sterols in tissue slices (10 µm) of mouse brain. The method provides sterolomic analysis at 400-µm spot diameter with a limit of quantification of 0.01 ng/mm2. It overcomes the limitations of previous mass spectrometry imaging techniques in analysis of low-abundance and difficult-to-ionize sterol molecules, allowing isomer differentiation and structure identification. Here we demonstrate the spatial distribution and quantification of multiple sterols involved in cholesterol metabolic pathways in wild-type and cholesterol 24S-hydroxylase knockout mouse brain. The technology described provides a powerful tool for future studies of spatial cholesterol metabolism in healthy and diseased tissues.
Collapse
|
21
|
Gamba P, Staurenghi E, Testa G, Giannelli S, Sottero B, Leonarduzzi G. A Crosstalk Between Brain Cholesterol Oxidation and Glucose Metabolism in Alzheimer's Disease. Front Neurosci 2019; 13:556. [PMID: 31213973 PMCID: PMC6554318 DOI: 10.3389/fnins.2019.00556] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 05/14/2019] [Indexed: 12/14/2022] Open
Abstract
In Alzheimer’s disease (AD), both cholesterol and glucose dysmetabolism precede the onset of memory deficit and contribute to the disease’s progression. It is indeed now believed that oxidized cholesterol in the form of oxysterols and altered glucose uptake are the main triggers in AD affecting production and clearance of Aβ, and tau phosphorylation. However, only a few studies highlight the relationship between them, suggesting the importance of further extensive studies on this topic. Recently, a molecular link was demonstrated between cholesterol oxidative metabolism and glucose uptake in the brain. In particular, 27-hydroxycholesterol, a key linker between hypercholesterolemia and the increased AD risk, is considered a biomarker for reduced glucose metabolism. In fact, its excess increases the activity of the renin-angiotensin system in the brain, thus reducing insulin-mediated glucose uptake, which has a major impact on brain functioning. Despite this important evidence regarding the role of 27-hydroxycholesterol in regulating glucose uptake by neurons, the involvement of other cholesterol oxidation products that have been clearly demonstrated to be key players in AD cannot be ruled out. This review highlights the current understanding of the potential role of cholesterol and glucose dysmetabolism in AD progression, and the bidirectional crosstalk between these two phenomena.
Collapse
Affiliation(s)
- Paola Gamba
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, Turin, Italy
| | - Erica Staurenghi
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, Turin, Italy
| | - Gabriella Testa
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, Turin, Italy
| | - Serena Giannelli
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, Turin, Italy
| | - Barbara Sottero
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, Turin, Italy
| | - Gabriella Leonarduzzi
- Department of Clinical and Biological Sciences, University of Turin, San Luigi Hospital, Turin, Italy
| |
Collapse
|
22
|
Björkhem I, Leoni V, Svenningsson P. On the fluxes of side-chain oxidized oxysterols across blood-brain and blood-CSF barriers and origin of these steroids in CSF (Review). J Steroid Biochem Mol Biol 2019; 188:86-89. [PMID: 30586624 DOI: 10.1016/j.jsbmb.2018.12.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 12/22/2018] [Indexed: 12/13/2022]
Abstract
In contrast to cholesterol itself the side-chain oxidized metabolites 24S-hydroxycholesterol (24OH) and 27-hydroxycholesterol (27OH) are able to pass the blood-brain barrier and the blood-CSF barrier. Most 27OH in circulation is formed extracerebrally and according to catheterization experiments about 5 mg of it is taken up by the brain per 24 h. 24OH is almost exclusively produced in the brain and about 6 mg fluxes from the brain into the circulation per 24 h. In addition to these major fluxes a very minor fraction of these two oxysterols flux from the circulation into CSF. Isotope experiments have shown that almost all 27OH in CSF originates from the circulation and evidence has been presented that this is the case also with a substantial part of 24OH. The levels of both 24OH and 27OH in CSF are thus affected by the integrity of the blood-CSF barrier with higher levels when the barrier is defect. Both levels of 24OH and 27OH in CSF are increased in connection with neurodegeneration and in general the increase in 24OH levels is higher than the increase in 27OH levels. A number of observations in different type of patients including measurements of other biochemical markers support that the increase in levels of 24OH due to neurodegeneration is due to a release of this oxysterol or its precursor cholesterol from dying neuronal cells. In contrast the increase in levels of 27OH is likely to be a consequence of reduced metabolism due to loss of the neuronal enzyme CYP7B1. We discuss the driving forces behind the fluxes of oxysterols in the brain, the limitations in the flux across the barriers and the diagnostic potential for side-chain oxidized oxysterols in CSF.
Collapse
Affiliation(s)
| | - Valerio Leoni
- Laboratory of Clinical Chemistry, Hospital of Varese, Varese, Italy
| | | |
Collapse
|
23
|
Oxysterols as a biomarker in diseases. Clin Chim Acta 2019; 491:103-113. [PMID: 30685361 DOI: 10.1016/j.cca.2019.01.022] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 01/20/2019] [Accepted: 01/23/2019] [Indexed: 12/24/2022]
Abstract
Cholesterol is one of the most important chemical substances as a structural element in human cells, and it is very susceptible to oxidation reactions that form oxysterol. Oxysterols exhibit almost the exact structure as cholesterol and a cholesterol precursor (7-dehydrocholesterol) with an additional hydroxyl, epoxy or ketone moiety. The oxidation reaction is performed via an enzymatic or non-enzymatic mechanism. The wide array of enzymatic oxysterols encountered in the human body varies in origin and function. Oxysterols establish a concentration equilibrium in human body fluids. Disease may alter the equilibrium, and oxysterols may be used as a diagnostic tool. The current review presents the possibility of using non-enzymatic oxysterols and disturbances in enzymatic oxysterol equilibrium in the human body as a potential biomarker for diagnosing and/or monitoring of the progression of various diseases.
Collapse
|
24
|
Abdel-Khalik J, Crick PJ, Yutuc E, DeBarber AE, Duell PB, Steiner RD, Laina I, Wang Y, Griffiths WJ. Identification of 7α,24-dihydroxy-3-oxocholest-4-en-26-oic and 7α,25-dihydroxy-3-oxocholest-4-en-26-oic acids in human cerebrospinal fluid and plasma. Biochimie 2018; 153:86-98. [PMID: 29960034 PMCID: PMC6171785 DOI: 10.1016/j.biochi.2018.06.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Accepted: 06/25/2018] [Indexed: 01/08/2023]
Abstract
Dihydroxyoxocholestenoic acids are intermediates in bile acid biosynthesis. Here, using liquid chromatography – mass spectrometry, we confirm the identification of 7α,24-dihydroxy-3-oxocholest-4-en-26-oic and 7α,25-dihydroxy-3-oxocholest-4-en-26-oic acids in cerebrospinal fluid (CSF) based on comparisons to authentic standards and of 7α,12α-dihydroxy-3-oxocholest-4-en-26-oic and 7α,x-dihydroxy-3-oxocholest-4-en-26-oic (where hydroxylation is likely on C-22 or C-23) based on exact mass measurement and multistage fragmentation. Surprisingly, patients suffering from the inborn error of metabolism cerebrotendinous xanthomatosis, where the enzyme CYP27A1, which normally introduces the (25 R)26-carboxylic acid group to the sterol side-chain, is defective still synthesise 7α,24-dihydroxy-3-oxocholest-4-en-26-oic acid and also both 25 R- and 25 S-epimers of 7α,12α-dihydroxy-3-oxocholest-4-en-26-oic acid. We speculate that the enzymes CYP46A1 and CYP3A4 may have C-26 carboxylase activity to generate these acids. In patients suffering from hereditary spastic paraplegia type 5 the CSF concentrations of the 7α,24- and 7α,25-dihydroxy acids are reduced, suggesting an involvement of CYP7B1 in their biosynthesis in brain. Dihydroxy-3-oxocholest-5-en-26-oic are found in human CSF and plasma. Hydroxy groups may be at 7α,24, 7α,25, or 7α,12α. Another acid is hydroxylated at 7α and in the side-chain probably at C-22 or C-23. In patients with CTX acids with 25 R or 25 S stereochemistry are found. In patients with SPG5 the concentrations of acids in CSF are reduced.
Collapse
Affiliation(s)
- Jonas Abdel-Khalik
- Institute of Life Science, Swansea University Medical School, ILS1 Building, Singleton Park, Swansea, SA2 8PP, UK
| | - Peter J Crick
- Institute of Life Science, Swansea University Medical School, ILS1 Building, Singleton Park, Swansea, SA2 8PP, UK
| | - Eylan Yutuc
- Institute of Life Science, Swansea University Medical School, ILS1 Building, Singleton Park, Swansea, SA2 8PP, UK
| | - Andrea E DeBarber
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, OR, USA
| | - P Barton Duell
- Knight Cardiovascular Institute, Oregon Health and Sciences University, Portland, OR, USA
| | - Robert D Steiner
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Ioanna Laina
- Athens Medical Group, Athens Medical Center, Marousi Athens, Greece
| | - Yuqin Wang
- Institute of Life Science, Swansea University Medical School, ILS1 Building, Singleton Park, Swansea, SA2 8PP, UK.
| | - William J Griffiths
- Institute of Life Science, Swansea University Medical School, ILS1 Building, Singleton Park, Swansea, SA2 8PP, UK.
| |
Collapse
|
25
|
Lütjohann D, Lopez AM, Chuang JC, Kerksiek A, Turley SD. Identification of Correlative Shifts in Indices of Brain Cholesterol Metabolism in the C57BL6/Mecp2 tm1.1Bird Mouse, a Model for Rett Syndrome. Lipids 2018; 53:363-373. [PMID: 29770459 DOI: 10.1002/lipd.12041] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 02/26/2018] [Accepted: 04/03/2018] [Indexed: 01/14/2023]
Abstract
Rett syndrome (RS) is a pervasive neurodevelopmental disorder resulting from loss-of-function mutations in the X-linked gene methyl-Cpg-binding protein 2 (MECP2). Using a well-defined model for RS, the C57BL6/Mecp2tm1.1Bird mouse, we have previously found a moderate but persistently lower rate of cholesterol synthesis, measured in vivo, in the brains of Mecp2-/y mice, starting from about the third week after birth. There was no genotypic difference in the total cholesterol concentration throughout the brain at any age. This raised the question of whether the lower rate of cholesterol synthesis in the mutants was balanced by a fall in the rate at which cholesterol was converted via cholesterol 24-hydroxylase (Cyp46A1) to 24S-hydroxycholesterol (24S-OHC), the principal route through which cholesterol is ordinarily removed from the brain. Here, we show that while there were no genotypic differences in the concentrations in plasma and liver of three cholesterol precursors (lanosterol, lathosterol, and desmosterol), two plant sterols (sitosterol and campesterol), and two oxysterols (27-hydroxycholesterol [27-OHC] and 24S-OHC), the brains of the Mecp2 -/y mice had significantly lower concentrations of all three cholesterol precursors, campesterol, and both oxysterols, with the level of 24S-OHC being ~20% less than in their Mecp2 +/y controls. Together, these data suggest that coordinated regulation of cholesterol synthesis and catabolism in the central nervous system is maintained in this model for RS. Furthermore, we speculate that the adaptive changes in these two pathways conceivably resulted from a shift in the permeability of the blood-brain barrier as implied by the significantly lower campesterol and 27-OHC concentrations in the brains of the Mecp2-/y mice.
Collapse
Affiliation(s)
- Dieter Lütjohann
- Institute for Clinical Chemistry and Clinical Pharmacology, University Clinics of Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
| | - Adam M Lopez
- Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9151, USA
| | - Jen-Chieh Chuang
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390-9060, USA
| | - Anja Kerksiek
- Institute for Clinical Chemistry and Clinical Pharmacology, University Clinics of Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
| | - Stephen D Turley
- Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9151, USA
| |
Collapse
|
26
|
Reinicke M, Schröter J, Müller-Klieser D, Helmschrodt C, Ceglarek U. Free oxysterols and bile acids including conjugates - Simultaneous quantification in human plasma and cerebrospinal fluid by liquid chromatography-tandem mass spectrometry. Anal Chim Acta 2018; 1037:245-255. [PMID: 30292299 DOI: 10.1016/j.aca.2018.02.049] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 02/14/2018] [Accepted: 02/18/2018] [Indexed: 01/19/2023]
Abstract
A liquid chromatography-electrospray ionization-tandem mass spectrometry (LC-ESI(+)-MS/MS) assay was developed and qualified for analyzing 35 analytes of the cholesterol metabolism, including free cholesterol, 17 free, non-esterified oxysterols and 17 free and conjugated bile acids in plasma and cerebrospinal fluid. As internal standards, 25 commercially available stable deuterium-labeled analogs of the analytes were used. Pre-analytical investigations included stability tests of analyte concentrations affected by different anticoagulation additives: lithium heparin-, citrate-, EDTA-K3-stabilized plasma and serum, and the stability in EDTA whole blood at RT. This LC-ESI(+)-MS/MS method was successfully applied for the analysis of paired serum/cerebrospinal fluid samples of patients with and without blood-brain barrier disturbance, as well as of 100 plasma samples of a LIFE-Adult study sub-cohort. A fast and simple sample preparation including protein precipitation and on-line solid-phase extraction was developed. As little as 55 μL of human plasma/serum or cerebrospinal fluid were needed for the analysis. It was possible to separate isomeric oxysterols and bile acids within 23 min using a C18 core-shell column. The assay is capable of quantifying in a linear range of 0.8-250 ng mL-1 for free hydroxycholesterols, 0.2-10 ng mL-1 for dihydroxycholesterols, 0.2-500 ng mL-1 for bile acids and 16-2000 μg mL-1 for cholesterol with acceptable accuracy and precision. In cerebrospinal fluid one free oxysterols, five free and five conjugated bile acids could be quantified. No significant differences between patients with and without blood-brain barrier disturbance were obtained. In the LIFE-Adult sub-cohort two free oxysterols, four free and seven conjugated bile acids could be quantified in EDTA plasma. Men showed significantly higher concentrations of 26-OHC than women (p = 0.035). Furthermore, in women lower levels of cholic acid, glycocholic acid, glycodeoxycholic acid, chenodeoxycholic acid, glycochenodeoxycholic acid, glycoursodeoxycholic acid, glycolithocholic acid and higher levels of taurocholic acid, taurochenodeoxycholic acid, ursodeoxycholic acid/hyodeoxycholic acid were quantified.
Collapse
Affiliation(s)
- Madlen Reinicke
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, Leipzig University, Liebigstr. 27, 04103 Leipzig, Germany.
| | - Jenny Schröter
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, Leipzig University, Liebigstr. 27, 04103 Leipzig, Germany
| | - Daniel Müller-Klieser
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, Leipzig University, Liebigstr. 27, 04103 Leipzig, Germany
| | - Christin Helmschrodt
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, Leipzig University, Liebigstr. 27, 04103 Leipzig, Germany
| | - Uta Ceglarek
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, Leipzig University, Liebigstr. 27, 04103 Leipzig, Germany; LIFE - Leipzig Research Center for Civilization Diseases, Leipzig University, Philipp-Rosenthal-Str. 27, 04103 Leipzig, Germany
| |
Collapse
|
27
|
Eggink HM, Tambyrajah LL, van den Berg R, Mol IM, van den Heuvel JK, Koehorst M, Groen AK, Boelen A, Kalsbeek A, Romijn JA, Rensen PCN, Kooijman S, Soeters MR. Chronic infusion of taurolithocholate into the brain increases fat oxidation in mice. J Endocrinol 2018; 236:85-97. [PMID: 29233934 DOI: 10.1530/joe-17-0503] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 12/11/2017] [Indexed: 01/11/2023]
Abstract
Bile acids can function in the postprandial state as circulating signaling molecules in the regulation of glucose and lipid metabolism via the transmembrane receptor TGR5 and nuclear receptor FXR. Both receptors are present in the central nervous system, but their function in the brain is unclear. Therefore, we investigated the effects of intracerebroventricular (i.c.v.) administration of taurolithocholate (tLCA), a strong TGR5 agonist, and GW4064, a synthetic FXR agonist, on energy metabolism. We determined the effects of chronic i.c.v. infusion of tLCA, GW4064, or vehicle on energy expenditure, body weight and composition as well as tissue specific fatty acid uptake in mice equipped with osmotic minipumps. We found that i.c.v. administration of tLCA (final concentration in cerebrospinal fluid: 1 μM) increased fat oxidation (tLCA group: 0.083 ± 0.006 vs control group: 0.036 ± 0.023 kcal/h, F = 5.46, P = 0.04) and decreased fat mass (after 9 days of tLCA infusion: 1.35 ± 0.13 vs controls: 1.96 ± 0.23 g, P = 0.03). These changes were associated with enhanced uptake of triglyceride-derived fatty acids by brown adipose tissue and with browning of subcutaneous white adipose tissue. I.c.v. administration of GW4064 (final concentration in cerebrospinal fluid: 10 μM) did not affect energy metabolism, body composition nor bile acid levels, negating a role of FXR in the central nervous system in metabolic control. In conclusion, bile acids such as tLCA may exert metabolic effects on fat metabolism via the brain.
Collapse
Affiliation(s)
- Hannah M Eggink
- Department of Endocrinology and MetabolismAcademic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
- Hypothalamic Integration MechanismsNetherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Lauren L Tambyrajah
- Division of EndocrinologyDepartment of Medicine, Leiden University Medical Centre, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical Centre, Leiden, The Netherlands
| | - Rosa van den Berg
- Division of EndocrinologyDepartment of Medicine, Leiden University Medical Centre, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical Centre, Leiden, The Netherlands
| | - Isabel M Mol
- Division of EndocrinologyDepartment of Medicine, Leiden University Medical Centre, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical Centre, Leiden, The Netherlands
| | - Jose K van den Heuvel
- Division of EndocrinologyDepartment of Medicine, Leiden University Medical Centre, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical Centre, Leiden, The Netherlands
| | - Martijn Koehorst
- Department of Pediatrics and Laboratory MedicineUniversity Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Albert K Groen
- Department of Pediatrics and Laboratory MedicineUniversity Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
- Department of Vascular MedicineAmsterdam Diabetes Centre, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Anita Boelen
- Department of Endocrinology and MetabolismAcademic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Andries Kalsbeek
- Department of Endocrinology and MetabolismAcademic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
- Hypothalamic Integration MechanismsNetherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Johannes A Romijn
- Department of MedicineAcademic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Patrick C N Rensen
- Division of EndocrinologyDepartment of Medicine, Leiden University Medical Centre, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical Centre, Leiden, The Netherlands
| | - Sander Kooijman
- Division of EndocrinologyDepartment of Medicine, Leiden University Medical Centre, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical Centre, Leiden, The Netherlands
| | - Maarten R Soeters
- Department of Endocrinology and MetabolismAcademic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
28
|
Abstract
Oxysterols are oxygenated derivatives of cholesterol formed in the human body or ingested in the diet. By modulating the activity of many proteins (for instance, liver X receptors, oxysterol-binding proteins, some ATP-binding cassette transporters), oxysterols can affect many cellular functions and influence various physiological processes (e.g., cholesterol metabolism, membrane fluidity regulation, intracellular signaling pathways). Due to their crucial role, it is important to be able to quantify them in pathological conditions. The method described here permits to measure the content of oxysterol in plasma, cell, or media using GC-MS.
Collapse
|
29
|
An Y, Zhang DD, Yu HL, Ma WW, Lu YH, Liu QR, Xiao R. 27-Hydroxycholesterol regulates cholesterol synthesis and transport in C6 glioma cells. Neurotoxicology 2017; 59:88-97. [PMID: 28167099 DOI: 10.1016/j.neuro.2017.02.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 02/01/2017] [Accepted: 02/02/2017] [Indexed: 01/19/2023]
Abstract
The oxysterol 27-Hydroxycholesterol (27-OHC) is a major cholesterol metabolite that can cross the blood brain barrier (BBB) from peripheral circulation to the brain. Currently, the role of 27-OHC on cholesterol homeostasis in astrocytes and the underlying mechanisms are not defined. Since all brain cholesterol is essentially synthesized in brain itself and astrocytes as net producers of cholesterol are essential for normal brain function, here we investigated the effects of 27-OHC on cholesterol synthesis and transport in C6 glioma cells. C6 cells were treated with 5, 10 and 20μM 27-OHC for 24h and the cell viability and apoptosis, the cholesterol levels and metabolism-related mediators, genes and proteins were subsequently assessed using cell-counting kit (CCK)-8, Amplex red, ELISA, real-time PCR and Western blot, respectively. We found that 27-OHC decreased cholesterol levels by down-regulating the expression of sterol-regulated element binding protein-1 (SREBP-1a), 3-hydroxy-3-methylglutaryl-CoA reductase (HMG-CR) and low density lipoprotein receptor (LDLR) and promoted cholesterol transport by up-regulating the expression of peroxisome proliferator-activated receptors-γ (PPAR-γ), liver X receptor-α (LXR-α), ATP-binding cassette transporter protein family member A1 (ABCA1) and apolipoprotein E (ApoE)genes. Our results suggested that 27-OHC may represent a sensitive modulator of cholesterol metabolism disorder by suppressing cholesterol synthesis and stimulating cholesterol transport in astrocytes.
Collapse
Affiliation(s)
- Yu An
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Dan-Di Zhang
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China; Department of Nutrition, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing 101149, China
| | - Huan-Ling Yu
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Wei-Wei Ma
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Yan-Hui Lu
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Quan-Ri Liu
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Rong Xiao
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
30
|
Griffiths WJ, Abdel-Khalik J, Yutuc E, Morgan AH, Gilmore I, Hearn T, Wang Y. Cholesterolomics: An update. Anal Biochem 2017; 524:56-67. [PMID: 28087213 PMCID: PMC5378159 DOI: 10.1016/j.ab.2017.01.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 12/23/2016] [Accepted: 01/09/2017] [Indexed: 01/04/2023]
Abstract
Cholesterolomics can be regarded as the identification and quantification of cholesterol, its precursors post squalene, and metabolites of cholesterol and of its precursors, in a biological sample. These molecules include 1,25-dihydroxyvitamin D3, steroid hormones and bile acids and intermediates in their respective biosynthetic pathways. In this short article we will concentrate our attention on intermediates in bile acid biosynthesis pathways, in particular oxysterols and cholestenoic acids. These molecular classes are implicated in the aetiology of a diverse array of diseases including autoimmune disease, Parkinson's disease, motor neuron disease, breast cancer, the lysosomal storage disease Niemann-Pick type C and the autosomal recessive disorder Smith-Lemli-Opitz syndrome. Mass spectrometry (MS) is the dominant technology for sterol analysis including both gas-chromatography (GC)-MS and liquid chromatography (LC)-MS and more recently matrix-assisted laser desorption/ionisation (MALDI)-MS for tissue imaging studies. Here we will discuss exciting biological findings and recent analytical improvements.
Collapse
Affiliation(s)
| | | | - Eylan Yutuc
- Swansea University Medical School, Singleton Park, Swansea SA2 8PP, UK
| | - Alwena H Morgan
- Swansea University Medical School, Singleton Park, Swansea SA2 8PP, UK
| | - Ian Gilmore
- Swansea University Medical School, Singleton Park, Swansea SA2 8PP, UK
| | - Thomas Hearn
- Swansea University Medical School, Singleton Park, Swansea SA2 8PP, UK
| | - Yuqin Wang
- Swansea University Medical School, Singleton Park, Swansea SA2 8PP, UK.
| |
Collapse
|
31
|
Saeed AA, Edström E, Pikuleva I, Eggertsen G, Björkhem I. On the importance of albumin binding for the flux of 7α-hydroxy-3-oxo-4-cholestenoic acid in the brain. J Lipid Res 2016; 58:455-459. [PMID: 27956474 DOI: 10.1194/jlr.p073403] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Indexed: 01/06/2023] Open
Abstract
We confirmed previous findings by a Japanese group that there is an accumulation of 7α-hydroxy-3-oxo-4-cholestenoic acid (7-Hoca) in human subdural hematomas. The accumulation correlated with the time from the bleeding to the sample collection. We present evidence that these accumulations are likely to be caused by the strong affinity of 7-Hoca to albumin and the marked difference between plasma and brain with respect to levels of albumin. In the circulation, 80-90% of 7-Hoca is bound to albumin with a ratio between the steroid acid and albumin of ∼1.4 ng/mg. In cerebrospinal fluid (CSF), the ratio between 7-Hoca and albumin is ∼30 ng/mg. When albumin or hemolyzed blood in a dialysis bag was exposed to CSF, there was a flux of 7-Hoca from CSF to the albumin. We suggest that the major explanation for accumulation of 7-Hoca in subdural hematoma is a flux from the brain into the hematoma due to the high affinity to albumin and the high capacity of 7-Hoca to pass biomembranes. We discuss the possibility that the markedly different ratios between 7-Hoca and albumin in circulation and brain can explain the flux of 7-Hoca from the brain into circulation against a concentration gradient.
Collapse
Affiliation(s)
- Ahmed A Saeed
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska University Hospital Huddinge, Karolinska Institute, Huddinge, Sweden.,Department of Biochemistry, Faculty of Medicine, University of Khartoum, Khartoum, Sudan
| | - Erik Edström
- Neurocentrum, Karolinska University Hospital Solna, Solna, Sweden
| | - Irina Pikuleva
- Neurocentrum, Karolinska University Hospital Solna, Solna, Sweden
| | - Gösta Eggertsen
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska University Hospital Huddinge, Karolinska Institute, Huddinge, Sweden
| | - Ingemar Björkhem
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska University Hospital Huddinge, Karolinska Institute, Huddinge, Sweden
| |
Collapse
|
32
|
Doria M, Maugest L, Moreau T, Lizard G, Vejux A. Contribution of cholesterol and oxysterols to the pathophysiology of Parkinson's disease. Free Radic Biol Med 2016; 101:393-400. [PMID: 27836779 DOI: 10.1016/j.freeradbiomed.2016.10.008] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 10/04/2016] [Accepted: 10/11/2016] [Indexed: 01/08/2023]
Abstract
Neurodegenerative diseases are a major public health issue worldwide. Some countries, including France, have engaged in research into the causes of Parkinson's disease, Alzheimer's disease, and multiple sclerosis and the management of these patients. It should lead to a better understanding of the mechanisms leading to these diseases including the possible involvement of lipids in their pathogenesis. Parkinson's disease is a progressive neurodegenerative disorder characterized by the loss of dopaminergic neurons in the substantia nigra and the accumulation of α-synuclein (Lewy bodies). Several in vivo studies have shown a relationship between the lipid profile [cholesterol, oxidized cholesterol products (oxysterols) formed either enzymatically or by auto-oxidation], the use of drugs regulating cholesterol levels, and the development of Parkinson's disease. Several oxysterols are present in the brain and could play a role in the development of this disease, particularly in the accumulation of α-synuclein, and through various side effects, such as oxidation, inflammation, and cell death. Consequently, in Parkinson's disease, some oxysterols could contribute to the pathophysiology of the disease and constitute potential biomarkers or therapeutic targets.
Collapse
Affiliation(s)
- Margaux Doria
- Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270, Université de Bourgogne Franche-Comté, INSERM, Dijon, France
| | - Lucie Maugest
- Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270, Université de Bourgogne Franche-Comté, INSERM, Dijon, France; Department of Neurology, University Hospital/University of Bourgogne Franche-Comté, Dijon, France
| | - Thibault Moreau
- Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270, Université de Bourgogne Franche-Comté, INSERM, Dijon, France; Department of Neurology, University Hospital/University of Bourgogne Franche-Comté, Dijon, France
| | - Gérard Lizard
- Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270, Université de Bourgogne Franche-Comté, INSERM, Dijon, France
| | - Anne Vejux
- Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270, Université de Bourgogne Franche-Comté, INSERM, Dijon, France.
| |
Collapse
|
33
|
Crick PJ, Griffiths WJ, Zhang J, Beibel M, Abdel-Khalik J, Kuhle J, Sailer AW, Wang Y. Reduced Plasma Levels of 25-Hydroxycholesterol and Increased Cerebrospinal Fluid Levels of Bile Acid Precursors in Multiple Sclerosis Patients. Mol Neurobiol 2016; 54:8009-8020. [PMID: 27878760 PMCID: PMC5684259 DOI: 10.1007/s12035-016-0281-9] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 10/31/2016] [Indexed: 12/18/2022]
Abstract
Multiple sclerosis (MS) is an autoimmune, inflammatory disease of the central nervous system (CNS). We have measured the levels of over 20 non-esterified sterols in plasma and cerebrospinal fluid (CSF) from patients suffering from MS, inflammatory CNS disease, neurodegenerative disease and control patients. Analysis was performed following enzyme-assisted derivatisation by liquid chromatography–mass spectrometry (LC–MS) exploiting multistage fragmentation (MSn). We found increased concentrations of bile acid precursors in CSF from each of the disease states and that patients with inflammatory CNS disease classified as suspected autoimmune disease or of unknown aetiology also showed elevated concentrations of 25-hydroxycholestertol (25-HC, P < 0.05) in CSF. Cholesterol concentrations in CSF were not changed except for patients diagnosed with amyotrophic lateral sclerosis (P < 0.01) or pathogen-based infections of the CNS (P < 0.05) where they were elevated. In plasma, we found that 25-HC (P < 0.01), (25R)26-hydroxycholesterol ((25R)26-HC, P < 0.05) and 7α-hydroxy-3-oxocholest-4-enoic acid (7αH,3O-CA, P < 0.05) were reduced in relapsing-remitting MS (RRMS) patients compared to controls. The pattern of reduced plasma levels of 25-HC, (25R)26-HC and 7αH,3O-CA was unique to RRMS. In summary, in plasma, we find that the concentration of 25-HC in RRMS patients is significantly lower than in controls. This is consistent with the hypothesis that a lower propensity of macrophages to synthesise 25-HC will result in reduced negative feedback by 25-HC on IL-1 family cytokine production and exacerbated MS. In CSF, we find that the dominating metabolites reflect the acidic pathway of bile acid biosynthesis and the elevated levels of these in CNS disease is likely to reflect cholesterol release as a result of demyelination or neuronal death. 25-HC is elevated in patients with inflammatory CNS disease probably as a consequence of up-regulation of the type 1 interferon-stimulated gene cholesterol 25-hydroxylase in macrophages.
Collapse
Affiliation(s)
- Peter J Crick
- Swansea University Medical School, Singleton Park, Swansea, SA2 8PP, UK
| | | | - Juan Zhang
- Analytical Science and Imaging, Novartis Institutes for BioMedical Research, CH-4002, Basel, Switzerland
| | - Martin Beibel
- Developmental & Molecular Pathways, Novartis Institutes for BioMedical Research, CH-4002, Basel, Switzerland
| | | | - Jens Kuhle
- Neurology, Departments of Medicine, Biomedicine and Clinical Research, University Hospital Basel, CH-4031, Basel, Switzerland
| | - Andreas W Sailer
- Developmental & Molecular Pathways, Novartis Institutes for BioMedical Research, CH-4002, Basel, Switzerland.
| | - Yuqin Wang
- Swansea University Medical School, Singleton Park, Swansea, SA2 8PP, UK.
| |
Collapse
|
34
|
Lopez AM, Chuang JC, Posey KS, Turley SD. Suppression of brain cholesterol synthesis in male Mecp2-deficient mice is age dependent and not accompanied by a concurrent change in the rate of fatty acid synthesis. Brain Res 2016; 1654:77-84. [PMID: 27789278 DOI: 10.1016/j.brainres.2016.10.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 10/13/2016] [Accepted: 10/21/2016] [Indexed: 12/29/2022]
Abstract
Mutations in the X-linked gene methyl-CpG-binding protein 2 (MECP2) are the principal cause of Rett syndrome, a progressive neurodevelopmental disorder afflicting 1 in 10,000 to 15,000 females. Studies using hemizygous Mecp2 mouse models have revealed disruptions to some aspects of their lipid metabolism including a partial suppression of cholesterol synthesis in the brains of mature Mecp2 mutants. The present studies investigated whether this suppression is evident from early neonatal life, or becomes manifest at a later stage of development. We measured the rate of cholesterol synthesis, in vivo, in the brains of male Mecp2-/y and their Mecp2+/y littermates at 7, 14, 21, 28, 42 and 56 days of age. Brain weight was consistently lower in the Mecp2-/y mice than in their Mecp2+/y controls except at 7 days of age. In the 7- and 14-day-old mice there was no genotypic difference in the rate of brain cholesterol synthesis but, from 21 days and later, it was always marginally lower in the Mecp2-/y mice than in age-matched Mecp2+/y littermates. At no age was a genotypic difference detected in either the rate of fatty acid synthesis or cholesterol concentration in the brain. Cholesterol synthesis rates in the liver and lungs of 56-day-old Mecp2-/y mice were normal. The onset of lower rates of brain cholesterol synthesis at about the time closure of the blood brain barrier purportedly occurs might signify a disruption to mechanism(s) that dictate intracellular levels of cholesterol metabolites including oxysterols known to exert a regulatory influence on the cholesterol biosynthetic pathway.
Collapse
Affiliation(s)
- Adam M Lopez
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Jen-Chieh Chuang
- Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Kenneth S Posey
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Stephen D Turley
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
35
|
Griffiths WJ, Abdel-Khalik J, Crick PJ, Yutuc E, Wang Y. New methods for analysis of oxysterols and related compounds by LC-MS. J Steroid Biochem Mol Biol 2016; 162:4-26. [PMID: 26639636 DOI: 10.1016/j.jsbmb.2015.11.017] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Revised: 11/23/2015] [Accepted: 11/25/2015] [Indexed: 12/21/2022]
Abstract
Oxysterols are oxygenated forms of cholesterol or its precursors. They are formed enzymatically and via reactive oxygen species. Oxysterols are intermediates in bile acid and steroid hormone biosynthetic pathways and are also bioactive molecules in their own right, being ligands to nuclear receptors and also regulators of the processing of steroid regulatory element-binding proteins (SREBPs) to their active forms as transcription factors regulating cholesterol and fatty acid biosynthesis. Oxysterols are implicated in the pathogenesis of multiple disease states ranging from atherosclerosis and cancer to multiple sclerosis and other neurodegenerative diseases including Alzheimer's and Parkinson's disease. Analysis of oxysterols is challenging on account of their low abundance in biological systems in comparison to cholesterol, and due to the propensity of cholesterol to undergo oxidation in air to generate oxysterols with the same structures as those present endogenously. In this article we review the mass spectrometry-based methods for oxysterol analysis paying particular attention to analysis by liquid chromatography-mass spectrometry (LC-MS).
Collapse
Affiliation(s)
- William J Griffiths
- College of Medicine, Grove Building, Swansea University, Singleton Park, Swansea SA2 8PP, UK.
| | - Jonas Abdel-Khalik
- College of Medicine, Grove Building, Swansea University, Singleton Park, Swansea SA2 8PP, UK
| | - Peter J Crick
- College of Medicine, Grove Building, Swansea University, Singleton Park, Swansea SA2 8PP, UK
| | - Eylan Yutuc
- College of Medicine, Grove Building, Swansea University, Singleton Park, Swansea SA2 8PP, UK
| | - Yuqin Wang
- College of Medicine, Grove Building, Swansea University, Singleton Park, Swansea SA2 8PP, UK.
| |
Collapse
|
36
|
Filomenko R, Fourgeux C, Bretillon L, Gambert-Nicot S. Oxysterols: Influence on plasma membrane rafts microdomains and development of ocular diseases. Steroids 2015; 99:259-65. [PMID: 25683893 DOI: 10.1016/j.steroids.2015.02.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 01/26/2015] [Accepted: 02/02/2015] [Indexed: 01/27/2023]
Abstract
Oxidation of cholesterol into oxysterols is a major way of elimination of cholesterol from the liver and extrahepatic tissues, including the brain and the retina. Oxysterols are involved in various cellular processes. Numerous links have been established between oxysterols and several disorders such as neurodegenerative pathologies, retinopathies and atherosclerosis. Different components of the lipid layer such as sphingolipids, sterols and proteins participate to membrane fluidity and forme lipid rafts microdomains. Few data are available on the links between lipids rafts and oxysterols. The purpose of this review is to suggest the potential role of lipid rafts microdomains in the development of retinopathies with special emphasis and opening perspectives of their interactions with oxysterols. Actually cholesterol oxidation mechanism may have deleterious effect on its ability to support rafts formation .This review suggest that the effect of oxysterols of lipid rafts would probably depend on the oxysterol molecule and cell type.
Collapse
Affiliation(s)
- Rodolphe Filomenko
- INRA, UMR1324 Centre des Sciences du Goût et de l'Alimentation, Equipe Œil, Nutrition et Signalisation Cellulaire, F-21000 Dijon, France; CNRS, UMR6265 Centre des Sciences du Goût et de l'Alimentation, F-21000 Dijon, France; Université de Bourgogne, Centre des Sciences du Goût et de l'Alimentation, F-21000 Dijon, France
| | - Cynthia Fourgeux
- INRA, UMR1324 Centre des Sciences du Goût et de l'Alimentation, Equipe Œil, Nutrition et Signalisation Cellulaire, F-21000 Dijon, France; CNRS, UMR6265 Centre des Sciences du Goût et de l'Alimentation, F-21000 Dijon, France; Université de Bourgogne, Centre des Sciences du Goût et de l'Alimentation, F-21000 Dijon, France
| | - Lionel Bretillon
- INRA, UMR1324 Centre des Sciences du Goût et de l'Alimentation, Equipe Œil, Nutrition et Signalisation Cellulaire, F-21000 Dijon, France; CNRS, UMR6265 Centre des Sciences du Goût et de l'Alimentation, F-21000 Dijon, France; Université de Bourgogne, Centre des Sciences du Goût et de l'Alimentation, F-21000 Dijon, France.
| | - Ségolène Gambert-Nicot
- INRA, UMR1324 Centre des Sciences du Goût et de l'Alimentation, Equipe Œil, Nutrition et Signalisation Cellulaire, F-21000 Dijon, France; CNRS, UMR6265 Centre des Sciences du Goût et de l'Alimentation, F-21000 Dijon, France; Université de Bourgogne, Centre des Sciences du Goût et de l'Alimentation, F-21000 Dijon, France; CHU Dijon, Service de Biochimie Clinique, F-21000 Dijon, France
| |
Collapse
|
37
|
Allinquant B, Clamagirand C, Potier MC. Role of cholesterol metabolism in the pathogenesis of Alzheimer's disease. Curr Opin Clin Nutr Metab Care 2014; 17:319-23. [PMID: 24839952 DOI: 10.1097/mco.0000000000000069] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW Cholesterol has been shown to stimulate the cleavage of amyloid precursor protein (APP) into amyloid peptides involved in Alzheimer's disease. However, high level of peripheral cholesterol as a risk factor for Alzheimer's disease is still debated. This current review provides an update of the recent literature on cholesterol and APP metabolisms in the brain. RECENT FINDINGS First, a new relationship between neuronal APP and cholesterol has been shown in which this protein controls cholesterol turnover required for neuronal activity. Second, oxysterols are able to stimulate the synthesis of ATP-binding cassette transporters involved in the exchange of amyloid peptides between the blood and the brain. Third, changes in APP targeting to lipid rafts and/or their composition in cholesterol regulate amyloid peptide production. SUMMARY These recent findings open new areas of investigations to control the neuronal activity and to decrease the amyloid peptide levels in brain, opening on new preventive and therapeutic strategies for Alzheimer's disease.
Collapse
Affiliation(s)
- Bernadette Allinquant
- aINSERM UMR 894, Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine bInstitut du Cerveau et de la Moelle, UPMC, INSERM UMR S 975, CNRS UMR 7225, Paris, France
| | | | | |
Collapse
|