1
|
Skubic C, Trček H, Nassib P, Kreft T, Walakira A, Pohar K, Petek S, Režen T, Ihan A, Rozman D. Knockouts of CYP51A1, DHCR24, or SC5D from cholesterol synthesis reveal pathways modulated by sterol intermediates. iScience 2024; 27:110651. [PMID: 39262789 PMCID: PMC11387598 DOI: 10.1016/j.isci.2024.110651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 05/20/2024] [Accepted: 07/31/2024] [Indexed: 09/13/2024] Open
Abstract
Sterols from cholesterol synthesis are crucial for cholesterol production, but also have individual roles difficult to assess in vivo due to essentiality of cholesterol. We developed HepG2 cell models with knockouts (KOs) for three enzymes of cholesterol synthesis, each accumulating specific sterols. Surprisingly, KOs of CYP51, DHCR24, and SC5D shared only 9% of differentially expressed genes. The most striking was the phenotype of CYP51 KO with highly elevated lanosterol and 24,25-dihydrolanosterol, significant increase in G2+M phase and enhanced cancer and cell cycle pathways. Comparisons with mouse liver Cyp51 KO data suggest 24,25-dihydrolanosterol activates similar cell proliferation pathways, possibly via elevated LEF1 and WNT/NFKB signaling. In contrast, SC5D and DHCR24 KO cells with elevated lathosterol or desmosterol proliferated slowly, with downregulated E2F, mitosis, and enriched HNF1A. These findings demonstrate that increase of lanosterol and 24,25-dihydrolanosterol, but not other sterols, promotes cell proliferation in hepatocytes.
Collapse
Affiliation(s)
- Cene Skubic
- Center for Functional Genomics and Bio-Chips, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Zaloška cesta 4, 1000 Ljubljana, Slovenia
| | - Hana Trček
- Center for Functional Genomics and Bio-Chips, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Zaloška cesta 4, 1000 Ljubljana, Slovenia
| | - Petra Nassib
- Center for Functional Genomics and Bio-Chips, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Zaloška cesta 4, 1000 Ljubljana, Slovenia
| | - Tinkara Kreft
- Center for Functional Genomics and Bio-Chips, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Zaloška cesta 4, 1000 Ljubljana, Slovenia
| | - Andrew Walakira
- Center for Functional Genomics and Bio-Chips, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Zaloška cesta 4, 1000 Ljubljana, Slovenia
| | - Katka Pohar
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Sara Petek
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Tadeja Režen
- Center for Functional Genomics and Bio-Chips, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Zaloška cesta 4, 1000 Ljubljana, Slovenia
| | - Alojz Ihan
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Damjana Rozman
- Center for Functional Genomics and Bio-Chips, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Zaloška cesta 4, 1000 Ljubljana, Slovenia
| |
Collapse
|
2
|
Rutkowska A, Eberl HC, Werner T, Hennrich ML, Sévin DC, Petretich M, Reddington JP, Pocha S, Gade S, Martinez-Segura A, Dvornikov D, Karpiak J, Sweetman GM, Fufezan C, Duempelfeld B, Braun F, Schofield C, Keles H, Alvarado D, Wang Z, Jansson KH, Faelth-Savitski M, Curry E, Remlinger K, Stronach EA, Feng B, Sharma G, Coleman K, Grandi P, Bantscheff M, Bergamini G. Synergistic Effects of PARP Inhibition and Cholesterol Biosynthesis Pathway Modulation. CANCER RESEARCH COMMUNICATIONS 2024; 4:2427-2443. [PMID: 39028932 PMCID: PMC11403291 DOI: 10.1158/2767-9764.crc-23-0549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 05/07/2024] [Accepted: 07/16/2024] [Indexed: 07/21/2024]
Abstract
An in-depth multiomic molecular characterization of PARP inhibitors revealed a distinct poly-pharmacology of niraparib (Zejula) mediated by its interaction with lanosterol synthase (LSS), which is not observed with other PARP inhibitors. Niraparib, in a similar way to the LSS inhibitor Ro-48-8071, induced activation of the 24,25-epoxysterol shunt pathway, which is a regulatory signaling branch of the cholesterol biosynthesis pathway. Interestingly, the combination of an LSS inhibitor with a PARP inhibitor that does not bind to LSS, such as olaparib, had an additive effect on killing cancer cells to levels comparable with niraparib as a single agent. In addition, the combination of PARP inhibitors and statins, inhibitors of 3-hydroxy-3-methylglutaryl-coenzyme A reductase, an enzyme catalyzing the rate-limiting step in the mevalonate pathway, had a synergistic effect on tumor cell killing in cell lines and patient-derived ovarian tumor organoids. These observations suggest that concomitant inhibition of the cholesterol biosynthesis pathway and PARP activity might result in stronger efficacy of these inhibitors against tumor types highly dependent on cholesterol metabolism. SIGNIFICANCE The presented data indicate, to our knowledge, for the first time, the potential benefit of concomitant modulation of cholesterol biosynthesis pathway and PARP inhibition and highlight the need for further investigation to assess its translational relevance.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Joel Karpiak
- Medicine Design-Computational Sciences, R&D, GSK, Heidelberg, Germany.
| | | | - Christian Fufezan
- Data Streams and Operations, and Data Science and Data Engineering, R&D, GSK, Heidelberg, Germany.
- Centre for Organismal Studies, Heidelberg University, Heidelberg, Germany.
| | | | - Florian Braun
- Chemical Biology Core Facility, EMBL Heidelberg, Heidelberg, Germany.
| | | | - Hakan Keles
- Genomic Sciences, R&D, GSK, Heidelberg, Germany.
| | - David Alvarado
- Oncology, Synthetic Lethality Research Unit, R&D, GSK, Heidelberg, Germany.
| | - Zhuo Wang
- Oncology, Synthetic Lethality Research Unit, R&D, GSK, Heidelberg, Germany.
| | | | | | - Edward Curry
- Genomic Sciences, R&D, GSK, Heidelberg, Germany.
| | | | | | - Bin Feng
- Oncology, Advanced Analytics Experimental Medicine Unit, R&D, GSK, Heidelberg, Germany.
| | - Geeta Sharma
- Oncology, Synthetic Lethality Research Unit, R&D, GSK, Heidelberg, Germany.
| | - Kevin Coleman
- Oncology, Synthetic Lethality Research Unit, R&D, GSK, Heidelberg, Germany.
| | | | | | | |
Collapse
|
3
|
Sun Z, Peng X, Zhao L, Yang Y, Zhu Y, Wang L, Kang B. From tissue lesions to neurotoxicity: The devastating effects of small-sized nanoplastics on red drum Sciaenops ocellatus. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 933:173238. [PMID: 38750760 DOI: 10.1016/j.scitotenv.2024.173238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/19/2024] [Accepted: 05/12/2024] [Indexed: 05/19/2024]
Abstract
Nanoplastic pollution typically exhibits more biotoxicity to marine organisms than microplastic pollution. Limited research exists on the toxic effects of small-sized nanoplastics on marine fish, especially regarding their post-exposure resilience. In this study, red drum (Sciaenops ocellatus) were exposed to small-sized polystyrene nanoplastics (30 nm, PS-NPs) for 7 days for the exposure experiments, followed by 14 days of recovery experiments. Histologically, hepatic lipid droplets and branchial epithelial liftings were the primary lesions induced by PS-NPs during both exposure and recovery periods. The inhibition of total superoxide dismutase activity and the accumulation of malondialdehyde content throughout the exposure and recovery periods. Transcriptional and metabolic regulation revealed that PS-NPs induced lipid metabolism disorders and DNA damage during the initial 1-2 days of exposure periods, followed by immune responses and neurotoxicity in the later stages (4-7 days). During the early recovery stages (2-7 days), lipid metabolism and cell cycle were activated, while in the later recovery stage (14 days), the emphasis shifted to lipid metabolism and energy metabolism. Persistent histological lesions, changes in antioxidant capacity, and fluctuations in gene and metabolite expression were observed even after 14 days of recovery periods, highlighting the severe biotoxicity of small-sized PS-NPs to marine fish. In summary, small-sized PS-NPs have severe biotoxicity, causing tissue lesions, oxidative damage, lipid metabolism disorders, DNA damage, immune responses, and neurotoxicity in red drum. This study offers valuable insights into the toxic effects and resilience of small-sized nanoplastics on marine fish.
Collapse
Affiliation(s)
- Zhicheng Sun
- Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao 266003, Shandong, China; Fisheries College, Ocean University of China, Qingdao 266003, Shandong, China
| | - Xin Peng
- Marine Academy of Zhejiang Province, Hangzhou 315613, Zhejiang, China; Key Laboratory of Ocean Space Resource Management Technology, Hangzhou 310012, Zhejiang, China
| | - Linlin Zhao
- Key Laboratory of Marine Eco-Environmental Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao 266061, Shandong, China
| | - Yi Yang
- State Key Laboratory of Marine Pollution and Department of Chemistry, City University of Hong Kong, 999077, Hong Kong, China
| | - Yugui Zhu
- Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao 266003, Shandong, China; Fisheries College, Ocean University of China, Qingdao 266003, Shandong, China
| | - Linlong Wang
- Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao 266003, Shandong, China; Fisheries College, Ocean University of China, Qingdao 266003, Shandong, China
| | - Bin Kang
- Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, Qingdao 266003, Shandong, China; Fisheries College, Ocean University of China, Qingdao 266003, Shandong, China
| |
Collapse
|
4
|
Wang HY, Nguyen TP, Sternisha AC, Carroll CL, Cross B, Morlock L, Williams NS, McBrayer S, Nijhawan D, De Brabander JK. Discovery and Optimization of N-Arylated Tetracyclic Dicarboximides That Target Primary Glioma Stem-like Cells. J Med Chem 2024; 67:9277-9301. [PMID: 38804887 DOI: 10.1021/acs.jmedchem.4c00402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
We recently discovered a novel N-aryl tetracyclic dicarboximide MM0299 (1) with robust activity against glioma stem-like cells that potently and selectively inhibits lanosterol synthase leading to the accumulation of the toxic shunt metabolite 24(S),25-epoxycholesterol. Herein, we delineate a systematic and comprehensive SAR study that explores the structural space surrounding the N-aryl tetracyclic dicarboximide scaffold. A series of 100 analogs were synthesized and evaluated for activity against the murine glioma stem-like cell line Mut6 and for metabolic stability in mouse liver S9 fractions. This study led to several analogs with single-digit nanomolar activity in Mut6 glioblastoma cells that were metabolically stable in S9 fractions. In vivo pharmacokinetic analysis of selected analogs identified compound 52a (IC50 = 63 nM; S9 T1/2 > 240 min) which was orally available (39% plasma; 58% brain) and displayed excellent brain exposure. Chronic oral dosing of 52a during a 2-week tolerability study indicated no adverse effect on body weight nor signs of hematologic, liver, or kidney toxicity.
Collapse
Affiliation(s)
- Hua-Yu Wang
- Department of Biochemistry, UT Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390, United States
| | - Thu P Nguyen
- Department of Biochemistry, UT Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390, United States
| | - Alex C Sternisha
- Children's Medical Center Research Institute and Department of Pediatrics, UT Southwestern, 5323 Harry Hines Boulevard, Dallas, Texas 75390, United States
| | - Christopher L Carroll
- Department of Biochemistry, UT Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390, United States
| | - Bethany Cross
- Department of Biochemistry, UT Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390, United States
| | - Lorraine Morlock
- Department of Biochemistry, UT Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390, United States
| | - Noelle S Williams
- Department of Biochemistry, UT Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390, United States
| | - Samuel McBrayer
- Children's Medical Center Research Institute and Department of Pediatrics, UT Southwestern, 5323 Harry Hines Boulevard, Dallas, Texas 75390, United States
| | - Deepak Nijhawan
- Department of Biochemistry, UT Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390, United States
- Department of Internal Medicine, Division of Hematology/Oncology and Program in Molecular Medicine, UT Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390, United States
| | - Jef K De Brabander
- Department of Biochemistry, UT Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390, United States
| |
Collapse
|
5
|
Peeples ES, Mirnics K, Korade Z. Chemical Inhibition of Sterol Biosynthesis. Biomolecules 2024; 14:410. [PMID: 38672427 PMCID: PMC11048061 DOI: 10.3390/biom14040410] [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: 02/25/2024] [Revised: 03/22/2024] [Accepted: 03/26/2024] [Indexed: 04/28/2024] Open
Abstract
Cholesterol is an essential molecule of life, and its synthesis can be inhibited by both genetic and nongenetic mechanisms. Hundreds of chemicals that we are exposed to in our daily lives can alter sterol biosynthesis. These also encompass various classes of FDA-approved medications, including (but not limited to) commonly used antipsychotic, antidepressant, antifungal, and cardiovascular medications. These medications can interfere with various enzymes of the post-lanosterol biosynthetic pathway, giving rise to complex biochemical changes throughout the body. The consequences of these short- and long-term homeostatic disruptions are mostly unknown. We performed a comprehensive review of the literature and built a catalogue of chemical agents capable of inhibiting post-lanosterol biosynthesis. This process identified significant gaps in existing knowledge, which fall into two main areas: mechanisms by which sterol biosynthesis is altered and consequences that arise from the inhibitions of the different steps in the sterol biosynthesis pathway. The outcome of our review also reinforced that sterol inhibition is an often-overlooked mechanism that can result in adverse consequences and that there is a need to develop new safety guidelines for the use of (novel and already approved) medications with sterol biosynthesis inhibiting side effects, especially during pregnancy.
Collapse
Affiliation(s)
- Eric S. Peeples
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE 68198, USA;
- Child Health Research Institute, Omaha, NE 68198, USA;
- Division of Neonatology, Children’s Nebraska, Omaha, NE 68114, USA
| | - Karoly Mirnics
- Child Health Research Institute, Omaha, NE 68198, USA;
- Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Pharmacology & Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Zeljka Korade
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE 68198, USA;
- Child Health Research Institute, Omaha, NE 68198, USA;
- Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
6
|
Guo J, Chen S, Zhang Y, Liu J, Jiang L, Hu L, Yao K, Yu Y, Chen X. Cholesterol metabolism: physiological regulation and diseases. MedComm (Beijing) 2024; 5:e476. [PMID: 38405060 PMCID: PMC10893558 DOI: 10.1002/mco2.476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 01/09/2024] [Accepted: 01/12/2024] [Indexed: 02/27/2024] Open
Abstract
Cholesterol homeostasis is crucial for cellular and systemic function. The disorder of cholesterol metabolism not only accelerates the onset of cardiovascular disease (CVD) but is also the fundamental cause of other ailments. The regulation of cholesterol metabolism in the human is an extremely complex process. Due to the dynamic balance between cholesterol synthesis, intake, efflux and storage, cholesterol metabolism generally remains secure. Disruption of any of these links is likely to have adverse effects on the body. At present, increasing evidence suggests that abnormal cholesterol metabolism is closely related to various systemic diseases. However, the exact mechanism by which cholesterol metabolism contributes to disease pathogenesis remains unclear, and there are still unknown factors. In this review, we outline the metabolic process of cholesterol in the human body, especially reverse cholesterol transport (RCT). Then, we discuss separately the impact of abnormal cholesterol metabolism on common diseases and potential therapeutic targets for each disease, including CVD, tumors, neurological diseases, and immune system diseases. At the end of this review, we focus on the effect of cholesterol metabolism on eye diseases. In short, we hope to provide more new ideas for the pathogenesis and treatment of diseases from the perspective of cholesterol.
Collapse
Affiliation(s)
- Jiarui Guo
- Eye Center of the Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang ProvinceChina
| | - Silong Chen
- Eye Center of the Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang ProvinceChina
| | - Ying Zhang
- Eye Center of the Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang ProvinceChina
- Institute of Translational MedicineZhejiang University School of MedicineHangzhouZhejiang ProvinceChina
| | - Jinxia Liu
- Eye Center of the Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang ProvinceChina
| | - Luyang Jiang
- Eye Center of the Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang ProvinceChina
| | - Lidan Hu
- National Clinical Research Center for Child HealthThe Children's HospitalZhejiang University School of MedicineHangzhouZhejiang ProvinceChina
| | - Ke Yao
- Eye Center of the Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang ProvinceChina
| | - Yibo Yu
- Eye Center of the Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang ProvinceChina
| | - Xiangjun Chen
- Eye Center of the Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang ProvinceChina
- Institute of Translational MedicineZhejiang University School of MedicineHangzhouZhejiang ProvinceChina
| |
Collapse
|
7
|
Abstract
All mammalian cell membranes contain cholesterol to maintain membrane integrity. The transport of this hydrophobic lipid is mediated by lipoproteins. Cholesterol is especially enriched in the brain, particularly in synaptic and myelin membranes. Aging involves changes in sterol metabolism in peripheral organs and also in the brain. Some of those alterations have the potential to promote or to counteract the development of neurodegenerative diseases during aging. Here, we summarize the current knowledge of general principles of sterol metabolism in humans and mice, the most widely used model organism in biomedical research. We discuss changes in sterol metabolism that occur in the aged brain and highlight recent developments in cell type-specific cholesterol metabolism in the fast-growing research field of aging and age-related diseases, focusing on Alzheimer's disease. We propose that cell type-specific cholesterol handling and the interplay between cell types critically influence age-related disease processes.
Collapse
Affiliation(s)
- Gesine Saher
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany;
| |
Collapse
|
8
|
Li W, Berlinicke C, Huang Y, Giera S, McGrath AG, Fang W, Chen C, Takaesu F, Chang X, Duan Y, Kumar D, Chang C, Mao HQ, Sheng G, Dodge JC, Ji H, Madden S, Zack DJ, Chamling X. High-throughput screening for myelination promoting compounds using human stem cell-derived oligodendrocyte progenitor cells. iScience 2023; 26:106156. [PMID: 36852281 PMCID: PMC9958491 DOI: 10.1016/j.isci.2023.106156] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/18/2022] [Accepted: 02/03/2023] [Indexed: 02/11/2023] Open
Abstract
Promoting myelination capacity of endogenous oligodendrocyte precursor cells (OPCs) is a promising therapeutic approach for CNS demyelinating disorders such as Multiple Sclerosis (MS). To aid in the discovery of myelination-promoting compounds, we generated a genome-engineered human pluripotent stem cell (hPSC) line that consists of three reporters: identification-and-purification tag, GFP, and secreted-NanoLuc, driven by the endogenous PDGFRA, PLP1, and MBP genes, respectively. Using this cell line, we established a high-throughput drug screening platform and performed a small-molecule screen, which identified at least two myelination-promoting small-molecule (Ro1138452 and SR2211) that target prostacyclin (IP) receptor and retinoic acid receptor-related orphan receptor γ (RORγ), respectively. Single-cell-transcriptomic analysis of differentiating OPCs treated with these molecules further confirmed that they promote oligodendrocyte differentiation and revealed several pathways that are potentially modulated by them. The molecules and their target pathways provide promising targets for the possible development of remyelination-based therapy for MS and other demyelinating disorders.
Collapse
Affiliation(s)
- Weifeng Li
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Cynthia Berlinicke
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Yinyin Huang
- Sanofi Inc., Translational Science, 350 Water Street, Cambridge, MA, 02141, USA
| | - Stefanie Giera
- Sanofi Inc., Rare and Neurologic Diseases Therapeutic Area, 350 Water Street, Cambridge, MA, 02141, USA
| | - Anna G. McGrath
- Sanofi Inc., Rare and Neurologic Diseases Therapeutic Area, 350 Water Street, Cambridge, MA, 02141, USA
| | - Weixiang Fang
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Chaoran Chen
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Felipe Takaesu
- Wallace H. Coulter Department of Biomedical Engineering, Emory University School of Medicine & Georgia Institute of Technology, Atlanta, GA, USA
| | - Xiaoli Chang
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Yukan Duan
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Dinesh Kumar
- Sanofi Inc., Translational Science, 350 Water Street, Cambridge, MA, 02141, USA
| | - Calvin Chang
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Hai-Quan Mao
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Whiting School of Engineering Baltimore, MD 21218, USA
| | - Guoqing Sheng
- Sanofi Inc., Rare and Neurologic Diseases Therapeutic Area, 350 Water Street, Cambridge, MA, 02141, USA
| | - James C. Dodge
- Sanofi Inc., Rare and Neurologic Diseases Therapeutic Area, 350 Water Street, Cambridge, MA, 02141, USA
| | - Hongkai Ji
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Stephen Madden
- Sanofi Inc., Translational Science, 350 Water Street, Cambridge, MA, 02141, USA
| | - Donald J. Zack
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Xitiz Chamling
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
9
|
Gao YH, Li X. Cholesterol metabolism: Towards a therapeutic approach for multiple sclerosis. Neurochem Int 2023; 164:105501. [PMID: 36803679 DOI: 10.1016/j.neuint.2023.105501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 01/26/2023] [Accepted: 01/30/2023] [Indexed: 02/17/2023]
Abstract
Growing evidence points to the importance of cholesterol in preserving brain homeostasis. Cholesterol makes up the main component of myelin in the brain, and myelin integrity is vital in demyelinating diseases such as multiple sclerosis. Because of the connection between myelin and cholesterol, the interest in cholesterol in the central nervous system increased during the last decade. In this review, we provide a detailed overview on brain cholesterol metabolism in multiple sclerosis and its role in promoting oligodendrocyte precursor cell differentiation and remyelination.
Collapse
Affiliation(s)
- Yu-Han Gao
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710119, China
| | - Xing Li
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710119, China.
| |
Collapse
|
10
|
Nguyen TP, Wang W, Sternisha AC, Corley CD, Wang HYL, Wang X, Ortiz F, Lim SK, Abdullah KG, Parada LF, Williams NS, McBrayer SK, McDonald JG, De Brabander JK, Nijhawan D. Selective and brain-penetrant lanosterol synthase inhibitors target glioma stem-like cells by inducing 24(S),25-epoxycholesterol production. Cell Chem Biol 2023; 30:214-229.e18. [PMID: 36758549 PMCID: PMC10008516 DOI: 10.1016/j.chembiol.2023.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 11/21/2022] [Accepted: 01/19/2023] [Indexed: 02/10/2023]
Abstract
Glioblastoma (GBM) is an aggressive adult brain cancer with few treatment options due in part to the challenges of identifying brain-penetrant drugs. Here, we investigated the mechanism of MM0299, a tetracyclic dicarboximide with anti-glioblastoma activity. MM0299 inhibits lanosterol synthase (LSS) and diverts sterol flux away from cholesterol into a "shunt" pathway that culminates in 24(S),25-epoxycholesterol (EPC). EPC synthesis following MM0299 treatment is both necessary and sufficient to block the growth of mouse and human glioma stem-like cells by depleting cellular cholesterol. MM0299 exhibits superior selectivity for LSS over other sterol biosynthetic enzymes. Critical for its application in the brain, we report an MM0299 derivative that is orally bioavailable, brain-penetrant, and induces the production of EPC in orthotopic GBM tumors but not normal mouse brain. These studies have implications for the development of an LSS inhibitor to treat GBM or other neurologic indications.
Collapse
Affiliation(s)
- Thu P Nguyen
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Wentian Wang
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Alex C Sternisha
- Children's Medical Center Research Institute and Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Chase D Corley
- Center for Human Nutrition, Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Hua-Yu Leo Wang
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xiaoyu Wang
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Francisco Ortiz
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sang-Kyun Lim
- Department of Development Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Kalil G Abdullah
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Luis F Parada
- Department of Development Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Noelle S Williams
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Samuel K McBrayer
- Children's Medical Center Research Institute and Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jeffrey G McDonald
- Center for Human Nutrition, Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jef K De Brabander
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Deepak Nijhawan
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Internal Medicine, Program in Molecular Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
11
|
The interaction between polyphyllin I and SQLE protein induces hepatotoxicity through SREBP-2/HMGCR/SQLE/LSS pathway. J Pharm Anal 2023; 13:39-54. [PMID: 36820075 PMCID: PMC9937801 DOI: 10.1016/j.jpha.2022.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 11/10/2022] [Accepted: 11/12/2022] [Indexed: 11/21/2022] Open
Abstract
Polyphyllin I (PPI) and polyphyllin II (PII) are the main active substances in the Paris polyphylla. However, liver toxicity of these compounds has impeded their clinical application and the potential hepatotoxicity mechanisms remain to be elucidated. In this work, we found that PPI and PII exposure could induce significant hepatotoxicity in human liver cell line L-02 and zebrafish in a dose-dependent manner. The results of the proteomic analysis in L-02 cells and transcriptome in zebrafish indicated that the hepatotoxicity of PPI and PII was associated with the cholesterol biosynthetic pathway disorders, which were alleviated by the cholesterol biosynthesis inhibitor lovastatin. Additionally, 3-hydroxy-3-methy-lglutaryl CoA reductase (HMGCR) and squalene epoxidase (SQLE), the two rate-limiting enzymes in the cholesterol synthesis, selected as the potential targets, were confirmed by the molecular docking, the overexpression, and knockdown of HMGCR or SQLE with siRNA. Finally, the pull-down and surface plasmon resonance technology revealed that PPI could directly bind with SQLE but not with HMGCR. Collectively, these data demonstrated that PPI-induced hepatotoxicity resulted from the direct binding with SQLE protein and impaired the sterol-regulatory element binding protein 2/HMGCR/SQLE/lanosterol synthase pathways, thus disturbing the cholesterol biosynthesis pathway. The findings of this research can contribute to a better understanding of the key role of SQLE as a potential target in drug-induced hepatotoxicity and provide a therapeutic strategy for the prevention of drug toxic effects with similar structures in the future.
Collapse
|
12
|
Molina-Gonzalez I, Miron VE, Antel JP. Chronic oligodendrocyte injury in central nervous system pathologies. Commun Biol 2022; 5:1274. [PMID: 36402839 PMCID: PMC9675815 DOI: 10.1038/s42003-022-04248-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 11/10/2022] [Indexed: 11/21/2022] Open
Abstract
Myelin, the membrane surrounding neuronal axons, is critical for central nervous system (CNS) function. Injury to myelin-forming oligodendrocytes (OL) in chronic neurological diseases (e.g. multiple sclerosis) ranges from sublethal to lethal, leading to OL dysfunction and myelin pathology, and consequent deleterious impacts on axonal health that drive clinical impairments. This is regulated by intrinsic factors such as heterogeneity and age, and extrinsic cellular and molecular interactions. Here, we discuss the responses of OLs to injury, and perspectives for therapeutic targeting. We put forward that targeting mature OL health in neurological disease is a promising therapeutic strategy to support CNS function.
Collapse
Affiliation(s)
- Irene Molina-Gonzalez
- grid.4305.20000 0004 1936 7988United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh, Scotland UK ,grid.4305.20000 0004 1936 7988Centre for Discovery Brain Sciences, Chancellor’s Building, The University of Edinburgh, Edinburgh, Scotland UK ,grid.4305.20000 0004 1936 7988Medical Research Council Centre for Reproductive Health, The Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, Scotland UK
| | - Veronique E. Miron
- grid.4305.20000 0004 1936 7988United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh, Scotland UK ,grid.4305.20000 0004 1936 7988Centre for Discovery Brain Sciences, Chancellor’s Building, The University of Edinburgh, Edinburgh, Scotland UK ,grid.4305.20000 0004 1936 7988Medical Research Council Centre for Reproductive Health, The Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, Scotland UK ,grid.415502.7Barlo Multiple Sclerosis Centre and Keenan Research Centre for Biomedical Science, Toronto, Canada ,grid.17063.330000 0001 2157 2938Department of Immunology, University of Toronto, Toronto, Canada
| | - Jack P. Antel
- grid.14709.3b0000 0004 1936 8649Neuroimmunology Unit, Montreal Neurological Institute, McGill University, Montreal, QC Canada
| |
Collapse
|
13
|
Caprariello AV, Adams DJ. The landscape of targets and lead molecules for remyelination. Nat Chem Biol 2022; 18:925-933. [PMID: 35995862 PMCID: PMC9773298 DOI: 10.1038/s41589-022-01115-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 07/18/2022] [Indexed: 12/24/2022]
Abstract
Remyelination, or the restoration of myelin sheaths around axons in the central nervous system, is a multi-stage repair process that remains a major need for millions of patients with multiple sclerosis and other diseases of myelin. Even into adulthood, rodents and humans can generate new myelin-producing oligodendrocytes, leading to the therapeutic hypothesis that enhancing remyelination could lessen disease burden in multiple sclerosis. Multiple labs have used phenotypic screening to identify dozens of drugs that enhance oligodendrocyte formation, and several hit molecules have now advanced to clinical evaluation. Target identification studies have revealed that a large majority of these hits share the ability to inhibit a narrow range of cholesterol pathway enzymes and thereby induce cellular accumulation of specific sterol precursors to cholesterol. This Perspective surveys the recent fruitful intersection of chemical biology and remyelination and suggests multiple approaches toward new targets and lead molecules to promote remyelination.
Collapse
Affiliation(s)
| | - Drew J Adams
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
| |
Collapse
|
14
|
Sax JL, Hershman SN, Hubler Z, Allimuthu D, Elitt MS, Bederman I, Adams DJ. Enhancers of Human and Rodent Oligodendrocyte Formation Predominantly Induce Cholesterol Precursor Accumulation. ACS Chem Biol 2022; 17:2188-2200. [PMID: 35833657 PMCID: PMC9773236 DOI: 10.1021/acschembio.2c00330] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Regeneration of myelin in the central nervous system is being pursued as a potential therapeutic approach for multiple sclerosis. Several labs have reported small molecules that promote oligodendrocyte formation and remyelination in vivo. Recently, we reported that many such molecules function by inhibiting a narrow window of enzymes in the cholesterol biosynthesis pathway. Here we describe a new high-throughput screen of 1,836 bioactive molecules and a thorough re-analysis of more than 60 molecules previously identified as promoting oligodendrocyte formation from human, rat, or mouse oligodendrocyte progenitor cells. These studies highlight that an overwhelming fraction of validated screening hits, including several molecules being evaluated clinically for remyelination, inhibit cholesterol pathway enzymes like emopamil-binding protein (EBP). To rationalize these findings, we suggest a model that relies on the high druggability of sterol-metabolizing enzymes and the ability of cationic amphiphiles to mimic the transition state of EBP. These studies further establish cholesterol pathway inhibition as a dominant mechanism among screening hits that enhance human, rat, or mouse oligodendrocyte formation.
Collapse
Affiliation(s)
- Joel L Sax
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Samantha N Hershman
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Zita Hubler
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Dharmaraja Allimuthu
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Matthew S Elitt
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Ilya Bederman
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Drew J Adams
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, United States
| |
Collapse
|
15
|
Daggubati V, Raleigh DR, Sever N. Sterol regulation of developmental and oncogenic Hedgehog signaling. Biochem Pharmacol 2022; 196:114647. [PMID: 34111427 PMCID: PMC8648856 DOI: 10.1016/j.bcp.2021.114647] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/02/2021] [Accepted: 06/04/2021] [Indexed: 02/03/2023]
Abstract
The Hedgehog (Hh) family of lipid-modified signaling proteins directs embryonic tissue patterning and postembryonic tissue homeostasis, and dysregulated Hh signaling drives familial and sporadic cancers. Hh ligands bind to and inhibit the tumor suppressor Patched and allow the oncoprotein Smoothened (SMO) to accumulate in cilia, which in turn activates the GLI family of transcription factors. Recent work has demonstrated that endogenous cholesterol and oxidized cholesterol derivatives (oxysterols) bind and modulate SMO activity. Here we discuss the myriad sterols that activate or inhibit the Hh pathway, with emphasis on endogenous 24(S),25-epoxycholesterol and 3β,5α-dihydroxycholest-7-en-6-one, and propose models of sterol regulation of SMO. Synthetic inhibitors of SMO have long been the focus of drug development efforts. Here, we discuss the possible utility of steroidal SMO ligands or inhibitors of enzymes involved in sterol metabolism as cancer therapeutics.
Collapse
Affiliation(s)
- Vikas Daggubati
- Departments of Radiation Oncology and Neurological Surgery, and Biomedical Sciences Graduate Program, University of California, San Francisco, CA, USA,Medical Scientist Training Program, University of California, San Francisco, CA, USA
| | - David R. Raleigh
- Departments of Radiation Oncology and Neurological Surgery, and Biomedical Sciences Graduate Program, University of California, San Francisco, CA, USA
| | - Navdar Sever
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA,Corresponding author: Navdar Sever, Department of Cell Biology, Harvard Medical School, 240 Longwood Avenue, LHRRB 405, Boston, MA 02115, USA, , Telephone: (617) 432-1612
| |
Collapse
|
16
|
Berghoff SA, Spieth L, Saher G. Local cholesterol metabolism orchestrates remyelination. Trends Neurosci 2022; 45:272-283. [DOI: 10.1016/j.tins.2022.01.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 01/07/2022] [Accepted: 01/21/2022] [Indexed: 12/19/2022]
|
17
|
Pleshinger MJ, Friedrich RM, Hubler Z, Rivera-León AM, Gao F, Yan D, Sax JL, Srinivasan R, Bederman I, Shick HE, Tesar PJ, Adams DJ. Inhibition of SC4MOL and HSD17B7 shifts cellular sterol composition and promotes oligodendrocyte formation. RSC Chem Biol 2022; 3:56-68. [PMID: 35128409 PMCID: PMC8729178 DOI: 10.1039/d1cb00145k] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 10/04/2021] [Indexed: 12/28/2022] Open
Abstract
While the cholesterol biosynthesis pathway has been extensively studied, recent work has forged new links between inhibition of specific sterol pathway enzymes, accumulation of their unique sterol substrates, and biological areas as diverse as cancer, immunology, and neurodegenerative disease. We recently reported that dozens of small molecules enhance formation of oligodendrocytes, a glial cell type lost in multiple sclerosis, by inhibiting CYP51, Sterol 14-reductase, or EBP and inducing cellular accumulation of their 8,9-unsaturated sterol substrates. Several adjacent pathway enzymes also have 8,9-unsaturated sterol substrates but have not yet been evaluated as potential targets for oligodendrocyte formation or in many other biological contexts, in part due to a lack of available small-molecule probes. Here, we show that genetic suppression of SC4MOL or HSD17B7 increases the formation of oligodendrocytes. Additionally, we have identified and optimized multiple potent new series of SC4MOL and HSD17B7 inhibitors and shown that these small molecules enhance oligodendrocyte formation. SC4MOL inhibitor CW4142 induced accumulation of SC4MOL's sterol substrates in mouse brain and represents an in vivo probe of SC4MOL activity. Mechanistically, the cellular accumulation of these 8,9-unsaturated sterols represents a central driver of enhanced oligodendrocyte formation, as exogenous addition of purified SC4MOL and HSD17B7 substrates but not their 8,9-saturated analogs promotes OPC differentiation. Our work validates SC4MOL and HSD17B7 as novel targets for promoting oligodendrocyte formation, underlines a broad role for 8,9-unsaturated sterols as enhancers of oligodendrocyte formation, and establishes the first high-quality small molecules targeting SC4MOL and HSD17B7 as novel tools for probing diverse areas of biology.
Collapse
Affiliation(s)
- Matthew J Pleshinger
- Department of Pharmacology, Case Western Reserve University School of Medicine Cleveland Ohio 44106 USA
| | - Ryan M Friedrich
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine Cleveland Ohio 44106 USA
| | - Zita Hubler
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine Cleveland Ohio 44106 USA
| | - Adrianna M Rivera-León
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine Cleveland Ohio 44106 USA
| | - Farrah Gao
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine Cleveland Ohio 44106 USA
| | - David Yan
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine Cleveland Ohio 44106 USA
| | - Joel L Sax
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine Cleveland Ohio 44106 USA
| | - Ramya Srinivasan
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine Cleveland Ohio 44106 USA
| | - Ilya Bederman
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine Cleveland Ohio 44106 USA
| | - H Elizabeth Shick
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine Cleveland Ohio 44106 USA
| | - Paul J Tesar
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine Cleveland Ohio 44106 USA
| | - Drew J Adams
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine Cleveland Ohio 44106 USA
| |
Collapse
|
18
|
Remyelination: what are the prospects for regenerative therapies in multiple sclerosis? Emerg Top Life Sci 2021; 5:705-709. [PMID: 34415022 DOI: 10.1042/etls20210164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/31/2021] [Accepted: 08/04/2021] [Indexed: 11/17/2022]
Abstract
Multiple sclerosis (MS) involves the immune system attacking the myelin sheaths surrounding axons and is a major cause of disability in working-age adults. Various approved therapies now provide reasonably good control over MS neuroinflammation, but none have a pronounced impact on the neurodegeneration associated with the disease. One prominent approach to fulfilling the unmet need for neuroprotective therapies, is the search for agents that promote 'remyelination', namely the generation of new oligodendrocytes that can form replacement myelin sheaths around denuded axons. In this article, I discuss some emerging targets for remyelinating therapies, mainly being pursued by recently formed small companies translating academic findings.
Collapse
|
19
|
Ghzaiel I, Sassi K, Zarrouk A, Nury T, Ksila M, Leoni V, Bouhaouala-Zahar B, Hammami S, Hammami M, Mackrill JJ, Samadi M, Ghrairi T, Vejux A, Lizard G. 7-Ketocholesterol: Effects on viral infections and hypothetical contribution in COVID-19. J Steroid Biochem Mol Biol 2021; 212:105939. [PMID: 34118414 PMCID: PMC8188774 DOI: 10.1016/j.jsbmb.2021.105939] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/04/2021] [Accepted: 06/07/2021] [Indexed: 02/06/2023]
Abstract
7-Ketocholesterol, which is one of the earliest cholesterol oxidization products identified, is essentially formed by the auto-oxidation of cholesterol. In the body, 7-ketocholesterol is both provided by food and produced endogenously. This pro-oxidant and pro-inflammatory molecule, which can activate apoptosis and autophagy at high concentrations, is an abundant component of oxidized Low Density Lipoproteins. 7-Ketocholesterol appears to significantly contribute to the development of age-related diseases (cardiovascular diseases, age-related macular degeneration, and Alzheimer's disease), chronic inflammatory bowel diseases and to certain cancers. Recent studies have also shown that 7-ketocholesterol has anti-viral activities, including on SARS-CoV-2, which are, however, lower than those of oxysterols resulting from the oxidation of cholesterol on the side chain. Furthermore, 7-ketocholesterol is increased in the serum of moderately and severely affected COVID-19 patients. In the case of COVID-19, it can be assumed that the antiviral activity of 7-ketocholesterol could be counterbalanced by its toxic effects, including pro-oxidant, pro-inflammatory and pro-coagulant activities that might promote the induction of cell death in alveolar cells. It is therefore suggested that this oxysterol might be involved in the pathophysiology of COVID-19 by contributing to the acute respiratory distress syndrome and promoting a deleterious, even fatal outcome. Thus, 7-ketocholesterol could possibly constitute a lipid biomarker of COVID-19 outcome and counteracting its toxic effects with adjuvant therapies might have beneficial effects in COVID-19 patients.
Collapse
Affiliation(s)
- Imen Ghzaiel
- University Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270 / Inserm, 21000 Dijon, France; University of Monastir, Faculty of Medicine, LR12ES05, Lab-NAFS 'Nutrition - Functional Food & Vascular Health', 5000 Monastir, Tunisia; University Tunis-El Manar, Faculty of Sciences of Tunis, 2092 Tunis, Tunisia.
| | - Khouloud Sassi
- University Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270 / Inserm, 21000 Dijon, France; University Tunis El Manar, Laboratory of Onco-Hematology (LR05ES05), Faculty of Medicine, 1007 Tunis, Tunisia.
| | - Amira Zarrouk
- University of Monastir, Faculty of Medicine, LR12ES05, Lab-NAFS 'Nutrition - Functional Food & Vascular Health', 5000 Monastir, Tunisia; University of Sousse, Faculty of Medicine, Sousse, Tunisia.
| | - Thomas Nury
- University Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270 / Inserm, 21000 Dijon, France.
| | - Mohamed Ksila
- University Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270 / Inserm, 21000 Dijon, France; University Tunis-El Manar, Loboratory of Neurophysiology, Cellular Physiopathology and Valorisation of BioMoleecules, LR18ES03, Department of Biologie, Faculty of Sciences, 2092 Tunis, Tunisia.
| | - Valerio Leoni
- Laboratory of Clinical Chemistry, Hospitals of Desio, ASST-Brianza and Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy.
| | - Balkiss Bouhaouala-Zahar
- Laboratory of Venoms and Therapeutic Molecules, Pasteur Institute of Tunis & University of Tunis El Manar, 1002 Tunis, Tunisia.
| | - Sonia Hammami
- University of Monastir, Faculty of Medicine, LR12ES05, Lab-NAFS 'Nutrition - Functional Food & Vascular Health', 5000 Monastir, Tunisia.
| | - Mohamed Hammami
- University of Monastir, Faculty of Medicine, LR12ES05, Lab-NAFS 'Nutrition - Functional Food & Vascular Health', 5000 Monastir, Tunisia.
| | - John J Mackrill
- Department of Physiology, School of Medicine, University College Cork, Cork, Ireland.
| | - Mohammad Samadi
- LCPMC-A2, ICPM, Dept of Chemistry, Univ. Lorraine, Metz Technopôle, Metz, France.
| | - Taoufik Ghrairi
- University Tunis-El Manar, Loboratory of Neurophysiology, Cellular Physiopathology and Valorisation of BioMoleecules, LR18ES03, Department of Biologie, Faculty of Sciences, 2092 Tunis, Tunisia.
| | - Anne Vejux
- University Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270 / Inserm, 21000 Dijon, France.
| | - Gérard Lizard
- University Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270 / Inserm, 21000 Dijon, France.
| |
Collapse
|